Sélection de la langue

Search

Sommaire du brevet 3068041 

Énoncé de désistement de responsabilité concernant l'information provenant de tiers

Une partie des informations de ce site Web a été fournie par des sources externes. Le gouvernement du Canada n'assume aucune responsabilité concernant la précision, l'actualité ou la fiabilité des informations fournies par les sources externes. Les utilisateurs qui désirent employer cette information devraient consulter directement la source des informations. Le contenu fourni par les sources externes n'est pas assujetti aux exigences sur les langues officielles, la protection des renseignements personnels et l'accessibilité.

Disponibilité de l'Abrégé et des Revendications

L'apparition de différences dans le texte et l'image des Revendications et de l'Abrégé dépend du moment auquel le document est publié. Les textes des Revendications et de l'Abrégé sont affichés :

  • lorsque la demande peut être examinée par le public;
  • lorsque le brevet est émis (délivrance).
(12) Brevet: (11) CA 3068041
(54) Titre français: PROCEDES AMELIORES DE MESURE DE NIVEAUX D'HYDROLASE A TERMINAISON CARBOXY D'UBIQUITINE L1 DANS LE SANG
(54) Titre anglais: IMPROVED METHODS FOR MEASURING UBIQUITIN CARBOXY-TERMINAL HYDROLASE L1 LEVELS IN BLOOD
Statut: Accordé et délivré
Données bibliographiques
(51) Classification internationale des brevets (CIB):
  • G1N 33/68 (2006.01)
(72) Inventeurs :
  • BELIGERE, GANGAMANI S. (Etats-Unis d'Amérique)
  • BRENNAN, MELISSA B. (Etats-Unis d'Amérique)
  • GRIESHABER, JESSICA (Etats-Unis d'Amérique)
  • PACENTI, DAVID (Etats-Unis d'Amérique)
  • DATWYLER, SAUL A. (Etats-Unis d'Amérique)
  • RAMP, JOHN (Etats-Unis d'Amérique)
(73) Titulaires :
  • ABBOTT LABORATORIES
(71) Demandeurs :
  • ABBOTT LABORATORIES (Etats-Unis d'Amérique)
(74) Agent: GOWLING WLG (CANADA) LLP
(74) Co-agent:
(45) Délivré: 2024-06-25
(86) Date de dépôt PCT: 2018-07-02
(87) Mise à la disponibilité du public: 2019-01-10
Requête d'examen: 2022-01-20
Licence disponible: S.O.
Cédé au domaine public: S.O.
(25) Langue des documents déposés: Anglais

Traité de coopération en matière de brevets (PCT): Oui
(86) Numéro de la demande PCT: PCT/US2018/040612
(87) Numéro de publication internationale PCT: US2018040612
(85) Entrée nationale: 2019-12-19

(30) Données de priorité de la demande:
Numéro de la demande Pays / territoire Date
62/528,187 (Etats-Unis d'Amérique) 2017-07-03

Abrégés

Abrégé français

L'invention concerne des procédés améliorés de traitement, de mesure et de détection de niveaux d'hydrolase à terminaison carboxy d'ubiquitine L1 (UCH-L1) dans des échantillons de sang prélevés chez un sujet humain à certains instants pendant environ 8 heures (ou environ 8 heures ou moins) après le prélèvement de l'échantillon du sujet. L'UCH-L1 est un biomarqueur précoce pour une lésion cérébrale traumatique (TBI), et il existe un besoin de procédés améliorés d'évaluation de l'UCH-L1 dans le sang, susceptibles de contribuer au diagnostic et à l'évaluation d'un sujet humain qui a subi, ou susceptible d'avoir subi, une lésion au niveau du cerveau.


Abrégé anglais

Disclosed herein are improved methods of processing, measuring, and detecting levels of ubiquitin carboxy-terminal hydrolase L1 (UCH-L1) in blood samples taken from a human subject at time points within about 8 hours (or about 8 hours or less) after obtaining the sample from the subject. UCH-L1 is an early biomarker for traumatic brain injury (TBI), and there is a need for improved methods for assessing UCH-L1 in blood can aid in the diagnosis and evaluation of a human subject who has sustained or may have sustained a head injury.

Revendications

Note : Les revendications sont présentées dans la langue officielle dans laquelle elles ont été soumises.


CLAIMS
What is claimed is:
1. A method of measuring an amount of ubiquitin carboxy-terminal hydrolase Ll
(UCH-L1)
in a whole blood sample obtained from a subject, wherein said method comprises
processing the sample within 4 hours, 6 hours, or 8 hours after the sample is
obtained from
the subject to avoid a rise in UCH-L1 level that results from storage of the
sample, wherein
processing the sample comprises:
a) performing a test that measures the amount of UCH-L1 in the sample;
b) separating plasma from blood cells in the sample; and, subsequently
performing a
test using the plasma that measures the amount of =UCH-Ll; or
c) separating serum from any clots that arise in the sample; and, subsequently
performing a test using the serum that measures the amount of UCH-L1,
thereby avoiding a rise in UCH-L1 levels that results from storage of the
sample for longer than 8
hours.
2. The method of claim 1, wherein the sample is obtained from the subject
using:
a) a container comprising an anticoagulant, wherein the anticoagulant is
heparin or
etheylenediaminetetraacetic acid (EDTA); or
b) a serum collection tube.
3. The method of claim 1 or claim 2, wherein the test is an immunoassay,
chemical analysis,
SDS PAGE and Western blot analysis, electrophoresis analysis, a protein assay,
a competitive
binding assay, a functional protein assay, chromatography, or
spectrophotometry.
4. The method of any one of claims 1-3, wherein the test is employed in a
clinical chemistry
format.
5. The method of claim 3 or claim 4 wherein the test is an immunoassay
comprising:
a) contacting the whole blood sample, plasma, or serum, either simultaneously
or
sequentially, in any order with:
122
Date Recue/Date Received 2023-06-29

(1) at least one capture antibody, which binds to an epitope on UCH-L1 or UCH-
L1 fragment to
form a capture antibody-UCH-L1 antigen complex, and
(2) at least one detection antibody which includes a detectable label and
binds to an epitope on
UCH-L1 that is not bound by the capture antibody, to form a UCH-L1 antigen-
detection antibody
complex,
such that a capture antibody-UCH-L1 antigen-detection antibody complex is
formed, and
b) measuring the amount or concentration of UCH-L1 in the whole
blood sample,
plasma, or serum based on a signal generated by the detectable label in the
capture antibody-UCH-L1 antigen-detection antibody complex.
6. The method of any one of claims 1-5, wherein the sample is maintained at
room
temperature for some period of time during the time point that the sample is
obtained from the
subject to a time point when the test is performed.
7. The method of any one of claims 1-5, wherein the sample is maintained at
a temperature
from 2 C to 8 C for some period of time during the time point that the
sample is obtained from
the subject to the time point when the test is performed.
8. The method of any one of claims 1-7, wherein the sample is not mixed
during the period
of time after the sample is obtained from the subject to the time point when
the test is performed.
9. The method of any one of claims 1-8, wherein the amount of UCH-L1 in the
sample from
the subject is assessed as a measure of traumatic brain injury, wherein the
subject may have
sustained an injury to the head.
10. A method of avoiding or preventing an increase or rise in ubiquitin
carboxy-terminal
hydrolase Ll (UCH-L1) levels between the period of time a whole blood sample
is obtained from
a subject and prior to performing an assay on the sample, the method
comprising processing the
sample within 4 hours, 6 hours, or 8 hours after the sample is obtained from
the subject to avoid
an increase or rise in UCH-L1 level that results from storage of the sample
prior to performing the
assay, wherein processing the sample comprises:
a) separating plasma from blood cells in the sample; and, subsequently
performing an
assay using the plasma that measures the amount of UCH-L1 in the sample; or
123
Date Recue/Date Received 2023-06-29

b) separating serum from any clots that arise in the sample; and, subsequently
performing an assay using the serum that measures the amount of UCH-L1 in the
sample
thereby avoiding or preventing an increase or rise in UCH-L1 levels that
results from storage of
the sample for longer than 8 hours.
11. The method of claim 10, wherein the sample is obtained from the subject
using:
a) a container comprising an anticoagulant, wherein the anticoagulant is
heparin or
EDTA; or
b) a serum collection tube.
12. The method of claim 10 or claim 11, wherein the assay is an
immunoassay, chemical
analysis, SDS PAGE and Western blot analysis, electrophoresis analysis, a
protein assay, a
competitive binding assay, a functional protein assay, chromatography, or
spectrophotometry.
13. The method of any one of claims 10-12, wherein the assay is employed in
a clinical
chemistry format.
14. The method of claim 12 or claim 13, wherein the assay is an immunoassay
comprising:
a) contacting the whole blood sample, plasma, or serum, either simultaneously
or
sequentially, in any order with:
(1) at least one capture antibody, which binds to an epitope on UCH-L1 or UCH-
L1 fragment to
fonn a capture antibody-UCH-L1 antigen complex, and
(2) at least one detection antibody which includes a detectable label and
binds to an epitope on
UCH-L1 that is not bound by the capture antibody, to form a UCH-L1 antigen-
detection antibody
complex,
such that a capture antibody-UCH-L1 antigen-detection antibody complex is
formed, and
b) measuring the amount or concentration of UCH-L1 in the whole blood sample,
plasma, or serum based on a signal generated by the detectable label in the
capture antibody-UCH-L1 antigen-detection antibody complex.
124
Date Recue/Date Received 2023-06-29

15. The method of any one of claims 10-14, wherein the sample is maintained
at room
temperature for some period of time between the time point that the sample is
obtained from the
subject and the time point when the assay is performed.
16. The method of any one of claims 10-14, wherein the sample is maintained
at a temperature
from 2 C to 8 C for some period of time between the time point that the
sample is obtained from
the subject and the time point when the assay is performed.
17. The method of any one of claims 10-16, wherein the sample is not mixed
between the
period of time between the time point that the sample is obtained from the
subject and the time
point when the test is performed.
18. The method of any one of claims 10-17, wherein the amount of UCH-L 1 in
the sample
from the subject is assessed as a measure of traumatic brain injury, wherein
the subject may have
sustained an injury to the head.
125
Date Recue/Date Received 2023-06-29

Description

Note : Les descriptions sont présentées dans la langue officielle dans laquelle elles ont été soumises.


IMPROVED METHODS FOR MEASURING UBIQUITIN CARBOXY-TERMINAL
HYDROLASE Li LEVELS IN BLOOD
RELATED APPLICATION INFORMATION
[0001] This application claims priority to U.S. 62/528,187 filed on July 3,
2017.
TECHNICAL FIELD
[0002] The present disclosure relates to improved methods of processing,
measuring, and
detecting levels of ubiquitin carboxy-terminal hydrolase Ll (UCH-L1) in blood
samples
taken from a human subject at time points within about 8 hours (or about 8
hours or less)
after obtaining the sample from the subject. UCH-L1 is an early biomarker for
traumatic
brain injury (TB!); therefore, improved methods for assessing UCH-L1 in blood
can aid in
the diagnosis and evaluation of a human subject who has sustained or may have
sustained a
head injury.
BACKGROUND
[0003] More than 5 million mild traumatic brain injuries (TBIs) occur each
year in the United
States alone. Clinical tools such as physical exams and questionnaires are
subjective and lack
sensitivity and specificity. Computerized tomography (CT) scan or magnetic
resonance
imaging (Mitt) are costly, not available in all parts of the world and are
routinely done only
if the clinical tools and/or questionnaire suggests that there is a need to
perform such
imaging. Therefore, high false negative rates have been associated with the
diagnosis and
evaluation of TBI. Clinicians and patients need objective, reliable
information to accurately
evaluate this condition to promote appropriate triage and recovery.
[0004] In some cases, biomarkers for FBI have been identified, including
ubiquitin carboxy-
terminal hydrolase Li (UCH-L1), lactate dehydrogenase (LDH), glial fibiillary
acid protein
(GFAP), neuron specific enolase (NSE), and s-loop, Although the use of
biomarker data is
promising for providing the objective and reliable diagnostic information
sought by
clinicians and patients, limited data have been available for the use of TBI
biomarkers in the
acute care setting to aid in patient evaluation and management.
1
Date Recue/Date Received 2023-06-29

[0005] Moreover, it has been challenging to determine the most advantageous
clinical
parameters and conditions for obtaining and processing patient samples, so
that the
specificity and accuracy provided by biomarker assessment can be established
and
maintained after obtaining samples from a patient. For example, it is well
known in the art
that preanalytical errors are quite frequent in the area of blood testing,
including point-of-care
testing ((See, Heyer, et al., Effectiveness of practices to reduce blood
sample hemolysis in
EDs: A laboratory medicine best practices systemic review and meta-analysis,
Cl/n.
Biochem. , 45(0):1012-1032 (September 2012)).
Unfortunately, approximately 60-80% of laboratory errors occur outside of
the analytical phase and most often outside of the laboratory (e.g., namely,
during the
preanalytical and post-analytical phase). Moreover, it is the preanalytical
phase that is most
vulnerable to errors. Some of the most common preanalytical errors include:
[0006] 1. Patient preparation errors such as sampling at the incorrect
time. Sampling time is
important because it is known that many analytes and blood parameters vary
with time
during the day, month or even seasons,
[0007] 2. Sampling techniques such as "milking" during capillary sample and
venous
sampling using a syringe. Excessive massage and squeezing around the puncture
site
(commonly called "milking") is often done when capillary blood flow is not
adequate to
obtain the necessary blood volume to fill the tube. Excessive massage can
cause falsely
decreased concentrations (up to 10%) of some analysis due to the dilution of
the blood
samples with tissue fluid and falsely increased concentration of potassium due
to sample
hemolysis. The use of syringes generally increases the risk of hemolysis and
clot formation.
[0008] 3. Underfilled or incorrectly filled collection tubes. When
collection tubes with
additives are used, care must be taken during sampling to fill the tubes until
the vacuum is
exhausted and blood flow ceases. It is important to ensure the proper blood-to-
additive ratio.
Underfilled tubes are one of the most common preanalytical errors. The
quantity of the
additive (e.g., anticoagulant) in tubes is such to ensure the proper ratio of
blood and
anticoagulant if the tube is completely filled.
[0009] 4. Hemolysis caused by blood lysis. Hemolysis is the most frequent
preanalytical
error. It is also the most common cause of sample rejection by a central
laboratory.
2
Date Recue/Date Received 2023-06-29

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
Hemolysis cases interference with many assays due to the release of the blood-
cell
components from the lysed blood cells. When blood samples are hemolyzed, a new
clinical
sample is often required.
100101 Thus, there is a need for improved methods of obtaining and
processing samples that
will reduce preanalytical errors thereby allowing for a more accurate
measurement of
biomarkers of TBI that can be used to diagnose and treat patients, especially
in the context of
acute care.
SUMMARY
100111 The present disclosure is directed to an improvement of a method of
measuring an
amount of ubiquitin carboxy-terminal hydrolase Li (UCH-L1) in a blood sample
obtained
from a subject. In one embodiment, the method includes processing the sample
from the
subject within no more than about eight hours (e.g., eight hours or less)
after the sample is
obtained from the subject, in order to avoid a rise in UCH-L1 level that
results from storage
of the sample. For example, in one aspect, the sample is processed within a
period of time
after the sample is obtained from the subject of from about zero hours to
about 6 hours. For
example, in another aspect, the sample is processed within a period of time
after the sample
is obtained from the subject of from about zero hours to about 2 hours. In yet
another
example, the sample is processed within a period of time after the sample is
obtained from
the subject of from about from about zero hours to about 1 hour. In still yet
another example,
the sample is processed within a period of time after the sample is obtained
from the subject
of from about 1 hour to about 8 hours. In still yet another example, the
sample is processed
within a period of time after the sample is obtained from the subject of from
about 1 hour to
about 6 hours. In still yet another example, the sample is processed within a
period of time
after the sample is obtained from the subject of from about 1 hour to about 2
hours. In still
yet another example, the sample is processed within a period of time after the
sample is
obtained from the subject of about 8 hours or less. In still yet another
example, the sample is
processed within a period of time after the sample is obtained from the
subject of about 6
hours or less. In still yet another example, the sample is processed within a
period of time
after the sample is obtained from the subject of about 4 hours or less. In
still yet another
3

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
example, the sample is processed within a period of time after the sample is
obtained from
the subject of about 2 hours or less. In still yet another example, the sample
is processed
within a period of time after the sample is obtained from the subject of about
1 hour or less.
[0012] In another aspect, the above described method further comprises
subsequently
performing an assay or test to measure the amount of UCH-L1 in the sample.
[0013] In yet another aspect, the above described method further comprises
subsequently
performing an assay or test to detect UCH-L1 in the sample.
[0014] In yet another aspect, in the above described method, the processing
of the sample
comprises separating plasma from blood cells in the sample and subsequently
performing an
assay using the plasma that measures the amount of UCH-Li in the sample.
[0015] In yet another aspect, in the above described method, the sample is
obtained from the
subject using a container comprising an anticoagulant selected from the group
consisting of
heparin and EDTA.
[0016] In yet another aspect, in the above described method, the processing
of the sample
comprises separating serum from any clots that arise in the sample and,
subsequently
performing an assay using the serum that measures the amount of UCH-L1 in the
sample.
[0017] In the above described method, the sample is collected using a serum
collection tube.
[0018] In the above described method, the assay comprises any method by which
UCH-L1
amount can be assessed (e.g, UCH-L1 is detected and/or measured). For example,
the assay
can be selected from the group consisting of an immunoassay, chemical
analysis, SDS PAGE
and Western blot analysis, electrophoresis analysis, a protein assay, a
competitive binding
assay, a functional protein assay, chromatography, and spectrophotometry. In
one aspect, the
assay employed is in a clinical chemistry format. In another aspect, the assay
employed is an
immunoassay comprising:
a) contacting the sample, either simultaneously or sequentially, in any order
with:
(1) at least one capture antibody, which binds to an epitope on UCH-L1 or UCH-
Li fragment to form a capture antibody-UCH-L1 antigen complex, and
(2) at least one detection antibody which includes a detectable label and
binds to
an epitope on UCH-L1 that is not bound by the capture antibody, to form a UCH-
Ll antigen-detection antibody complex,
4

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
such that a capture antibody-UCH-L1 antigen-detection antibody complex is
formed, and
b) measuring the amount or concentration of UCH-Li in the sample based on the
signal generated by the detectable label in the capture antibody-UCH-L1
antigen-
detection antibody complex.
[0019] In yet another aspect, in the above described method, the sample is
maintained at room
temperature for some period of time between the time point that the sample is
obtained from
the subject and the time point when the assay is performed.
[0020] In yet another aspect, in the above described method, the sample is
maintained at a
temperature from about 2 C to about 8 C for some period of time between the
time point
that the sample is obtained from the subject and the time point when the assay
is performed.
[0021] In yet another aspect, in the above described method, the sample is
not mixed during
or between the period of time between the time point that the sample is
obtained from the
subject and the time point when the assay is performed.
[0022] In yet another aspect, in the above described method, the UCH-L1
amount in the
sample from the subject is assessed as a measure of traumatic brain injury,
wherein the
subject has sustained or may have sustained an injury to the head.
[0023] In another embodiment, the present invention is directed to a method
of avoiding or
preventing an increase or rise in UCH-Li levels between the period of time a
whole blood
sample is obtained from a subject and prior to performing an assay on the
sample (such as,
for example to detected and/or measure the amount of UCH-L1 in the sample).
Specifically,
the method avoids or prevents an erroneously high level of UCH-L1 in the
sample between
the period of time the sample is obtained from the subject and prior to
performing the assay,
which can lead to false positives, erroneous or other mistaken results. The
method involves
the step of processing the sample within no more than about eight hours (such
as, for
example, 8 hours or less) after the sample is obtained from the subject to
avoid an increase or
rise in UCH-L1 level that results from storage of the sample prior to
performing the assay.
For example, in one aspect, the sample is processed within a period of time
after the sample
is obtained from the subject of from about zero hours to about 6 hours. For
example, in
another aspect, the sample is processed within a period of time after the
sample is obtained

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
from the subject of from about zero hours to about 2 hours. In yet another
example, the
sample is processed within a period of time after the sample is obtained from
the subject of
from about from about zero hours to about 1 hour. In still yet another
example, the sample is
processed within a period of time after the sample is obtained from the
subject of from about
1 hour to about 8 hours. In still yet another example, the sample is processed
within a period
of time after the sample is obtained from the subject of from about 1 hour to
about 6 hours.
In still yet another example, the sample is processed within a period of time
after the sample
is obtained from the subject of from about 1 hour to about 2 hours. In still
yet another
example, the sample is processed within a period of time after the sample is
obtained from
the subject of about 8 hours or less. In still yet another example, the sample
is processed
within a period of time after the sample is obtained from the subject of about
6 hours or less.
In still yet another example, the sample is processed within a period of time
after the sample
is obtained from the subject of about 4 hours or less. In still yet another
example, the sample
is processed within a period of time after the sample is obtained from the
subject of about 2
hours or less. In still yet another example, the sample is processed within a
period of time
after the sample is obtained from the subject of about 1 hour or less.
[0024] In another aspect, the above described method further comprises
subsequently
performing an assay or test to measure the amount of UCH-Li in the sample.
[0025] In yet another aspect, the above described method further comprises
subsequently
performing an assay or test to detect UCH-L1 in the sample.
[0026] In yet another aspect, in the above described method, the processing
of the sample
comprises separating plasma from blood cells in the sample and subsequently
performing an
assay using the plasma that measures the amount of UCH-L1 in the sample.
[0027] In yet another aspect, in the above described method, the sample is
obtained from the
subject using a container comprising an anticoagulant selected from the group
consisting of
heparin and EDTA.
[0028] In yet another aspect, in the above described method, the processing
of the sample
comprises separating serum from any clots that arise in the sample and,
subsequently
performing an assay using the serum that measures the amount of UCH-L1 in the
sample.
[0029] In the above described method, the sample is collected using a serum
collection tube.
6

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0030] In the above described method, the assay comprises any method by which
UCH-L1
amount can be assessed (e.g, UCH-L1 is detected and/or measured). For example,
the assay
can be selected from the group consisting of an immunoassay, chemical
analysis, SDS PAGE
and Western blot analysis, electrophoresis analysis, a protein assay, a
competitive binding
assay, a functional protein assay, chromatography, and spectrophotometry. In
one aspect, the
assay employed is in a clinical chemistry format. In another aspect, the assay
employed is an
immunoassay comprising:
a) contacting the sample, either simultaneously or sequentially, in any order
with:
(1) at least one capture antibody, which binds to an epitope on UCH-L1 or UCH-
Li fragment to form a capture antibody-UCH-L1 antigen complex, and
(2) at least one detection antibody which includes a detectable label and
binds to
an epitope on UCH-L1 that is not bound by the capture antibody, to form a UCH-
Li antigen-detection antibody complex,
such that a capture antibody-UCH-L1 antigen-detection antibody complex is
formed, and
b) measuring the amount or concentration of UCH-L1 in the sample based on the
signal generated by the detectable label in the capture antibody-UCH-L1
antigen-
detection antibody complex.
[0031] In yet another aspect, in the above described method, the sample is
maintained at room
temperature for some period of time between the time point that the sample is
obtained from
the subject and the time point when the assay is performed.
[0032] In yet another aspect, in the above described method, the sample is
maintained at a
temperature from about 2 C to about 8 C for some period of time between the
time point
that the sample is obtained from the subject and the time point when the assay
is performed.
[0033] In yet another aspect, in the above described method, the sample is
not mixed during
or between the period of time between the time point that the sample is
obtained from the
subject and the time point when the assay is performed.
[0034] In yet another aspect, in the above described method, the UCH-L1
amount in the
sample from the subject is assessed as a measure of traumatic brain injury,
wherein the
subject has sustained or may have sustained an injury to the head.
7

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
BRIEF DESCRIPTION OF THE DRAWINGS
[0035] FIGS. 1A-1B show representative bar graphs of UCH-L1 levels in whole
blood
samples.
[0036] FIGS. 2A-2B show representative bar graphs of UCH-L1 levels in plasma
samples.
[0037] FIGS. 3A-3B show representative bar graphs of UCH-L1 levels in whole
blood
samples obtained using lithium heparin (FIG. 3A) or EDTA (FIG. 3B) collection
tubes.
[0038] FIGS. 4A-4B show representative bar graphs of UCH-L1 levels in plasma
samples
obtained using with EDTA collection tubes at room temperature (FIG. 4A) or 2-8
C (FIG.
4B).
[0039] FIGS. 5A-5B show representative bar graphs of UCH-L1 levels in
plasma samples
obtained using with lithium heparin collection tubes at room temperature (FIG.
5A) or 2-8 C
(FIG. 5B).
[0040] FIGS. 6A-6B show representative bar graphs of UCH-L1 levels in serum
samples at
room temperature (FIG. 6A) or 2-8 C (FIG. 6B).
[0041] FIG. 7 shows a representative bar graph of UCH-L1 levels in serum
samples with or
without exposure to a blood clot.
[0042] FIG. 8 shows a representative line graph comparing hemoglobin (Hgb)
levels to UCH-
Li levels in hemolysate samples spiked with plasma from EDTA collection tubes.
[0043] FIG. 9 shows a representative bar graph of UCH-L1 levels in whole
blood samples
obtained using EDTA collection tubes spiked with UCH-L1 recombinant analyte.
DETAILED DESCRIPTION
[0044] The present disclosure relates to improved methods of processing,
measuring, and
detecting levels of ubiquitin carboxy-terminal hydrolase Li (UCH-L1) in blood
samples
taken from a human subject at time points within about 8 hours or about 8
hours or less after
obtaining the sample from the subject. The methods disclosed herein can be
useful for
assessing UCH-L1 (generally considered a biomarker of TBI) in blood to aid in
the diagnosis
and evaluation of a human subject who has sustained or may have sustained a
head injury.
Additionally, studies have reported inconsistent UCH-L1 levels in blood over
the first 24
hour period after the blood samples have been obtained from a subject, and at
least part of
8

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
this increase may be due to preanalytical processing conditions or factors
that take place or
occur prior to the time that the samples are tested or an assay is performed
(e.g., to detect
and/or measure UCH-Li levels). For example, it has been reported that certain
standard
processing conditions that cause hemolysis have little to no effect on UCH-L1
levels.
However, the present disclosure has surprisingly found that certain
preanalytical processing
conditions and methods can cause an increase (namely, an erroneous increase)
in UCH-L1
levels in whole blood, thus resulting in false positives; such false positives
can undermine
thge accuracy of using UCH-L1 as a diagnostic marker for aiding in the
detection,
determination and/or assessment of disorders such as TBI.
[0045] Thus, the present disclosure provides methods to improve the manner
in which UCH-
Li can be obtained and measured in samples, such as in blood samples (e.g.,
whole blood or
plasma). Specifically, the present disclosure provides methods of avoiding or
preventing an
increase or rise in ubiquitin carboxy-terminal hydrolase Li (UCH-L1) level in
a blood
sample (e.g., whole blood sample or a plasma sample) obtained from a subject
prior to
performing a test or an assay. More specifically, methods described herein
avoid or prevent
an erroneously high level of UCH-L1 in the sample between the period of time
the sample is
obtained from the subject and prior to performing the assay. Such methods
involve
processing a blood sample within no more than about eight hours after the
sample is obtained
from the subject to avoid an increase or rise in UCH-L1 level that results
from storing the
sample prior to performing the assay. Such methods are effective to stabilize
the levels of
UCH-L1 in the blood sample until such time as a test or assay is performed to
assess (e.g.,
detect and/or measure) the UCH-L1 in the sample.
[0046] Section headings as used in this section and the entire disclosure
herein are merely for
organizational purposes and are not intended to be limiting.
1. Definitions
[0047] Unless otherwise defined, all technical and scientific terms used
herein have the same
meaning as commonly understood by one of ordinary skill in the art. In case of
conflict, the
present document, including definitions, will control. Preferred methods and
materials are
described below, although methods and materials similar or equivalent to those
described
9

herein can be used in practice or testing of the present disclosure.
The materials, methods, and examples disclosed herein are illustrative only
and not intended to be limiting.
[0048] The terms "comprise(s)," "include(s)," "having," "has," "can,"
"contain(s)," and
variants thereof, as used herein, are intended to be open-ended transitional
phrases, terms, or
words that do not preclude the possibility of additional acts or structures.
The singular forms
"a," "an" and "the" include plural references unless the context clearly
dictates otherwise.
The present disclosure also contemplates other embodiments "comprising,"
"consisting of'
and "consisting essentially of," the embodiments or elements presented herein,
whether
explicitly set forth or not.
[0049] For the recitation of numeric ranges herein, each intervening number
there between
with the same degree of precision is explicitly contemplated. For example, for
the range of
6-9, the numbers 7 and 8 are contemplated in addition to 6 and 9, and for the
range 6.0-7,0,
the number 6.0, 6.1, 6.2, 6,3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, and 7.0 are
explicitly contemplated.
[0050] "Antibody" and "antibodies" as used herein refers to monoclonal
antibodies,
multispecific antibodies, human antibodies, humanized antibodies (fully or
partially
humanized), animal antibodies such as, but not limited to, a bird (for
example, a duck or a
goose), a shark, a whale, and a mammal, including a non-primate (for example,
a cow, a pig,
a camel, a llama, a horse, a goat, a rabbit, a sheep, a hamster, a guinea pig,
a cat, a dog, a rat,
a mouse, etc.) or a non-human primate (for example, a monkey, a chimpanzee,
etc.),
recombinant antibodies, chimeric antibodies, single-chain Fvs ("scFv"), single
chain
antibodies, single domain antibodies, Fab fragments, F(ab') fragments, F(ab')2
fragments,
disulfide-linked Fvs ("sdFv"), and anti-idiotypic ("anti-Id") antibodies, dual-
domain
antibodies, dual variable domain (DVD) or triple variable domain (TVD)
antibodies (dual-
variable domain immunoglobulins and methods for making them are described in
Wu, C., et
al., Nature Biotechnology, 25(11):1290-1297 (2007) and PCT International
Application WO
2001/058956), and
functionally active epitope-binding fragments of any of the above. In
particular, antibodies
include immunoglobulin molecules and immunologically active fragments of
Date Recue/Date Received 2023-06-29

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
immunoglobulin molecules, namely, molecules that contain an analyte-binding
site.
Immunoglobulin molecules can be of any type (for example, IgG, IgE, IgM, IgD,
IgA, and
IgY), class (for example, IgGl, IgG2, IgG3, IgG4, IgAl, and IgA2), or
subclass. For
simplicity sake, an antibody against an analyte is frequently referred to
herein as being either
an "anti-analyte antibody" or merely an "analyte antibody" (e.g., an anti-UCH-
L1 antibody
or a UCH-L1 antibody).
[0051] "Affinity matured antibody" is used herein to refer to an antibody
with one or more
alterations in one or more CDRs, which result in an improvement in the
affinity (i.e., KD, kd
or ka) of the antibody for a target antigen compared to a parent antibody,
which does not
possess the alteration(s). Exemplary affinity matured antibodies will have
nanomolar or even
picomolar affinities for the target antigen. A variety of procedures for
producing affinity
matured antibodies is known in the art, including the screening of a
combinatory antibody
library that has been prepared using bio-display. For example, Marks et al.,
BioTechnology,
10: 779-783 (1992) describes affinity maturation by VH and VL domain
shuffling. Random
mutagenesis of CDR and/or framework residues is described by Barbas et al.,
Proc. Nat.
Acad. Sci. USA, 91: 3809-3813 (1994); Schier et al., Gene, 169: 147-155
(1995); Yelton et
al., J. Immunol., 155: 1994-2004 (1995); Jackson et al., J. Immunol., 154(7):
3310-3319
(1995); and Hawkins et al, J. Mol. Biol., 226: 889-896 (1992). Selective
mutation at
selective mutagenesis positions and at contact or hypermutation positions with
an activity-
enhancing amino acid residue is described in U.S. Patent No. 6,914,128 Bl.
[0052] "Antibody fragment" as used herein refers to a portion of an intact
antibody
comprising the antigen-binding site or variable region. The portion does not
include the
constant heavy chain domains (i.e., CH2, CH3, or CH4, depending on the
antibody isotype)
of the Fc region of the intact antibody. Examples of antibody fragments
include, but are not
limited to, Fab fragments, Fab' fragments, Fab'-SH fragments, F(abi)2
fragments, Fd
fragments, Fv fragments, diabodies, single-chain Fv (scFv) molecules, single-
chain
polypeptides containing only one light chain variable domain, single-chain
polypeptides
containing the three CDRs of the light-chain variable domain, single-chain
polypeptides
containing only one heavy chain variable region, and single-chain polypeptides
containing
the three CDRs of the heavy chain variable region.
11

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0053] The "area under curve" or "AUC" refers to area under a ROC curve. AUC
under a
ROC curve is a measure of accuracy. An AUC of 1 represents a perfect test,
whereas an
AUC of 0.5 represents an insignificant test. A preferred AUC may be at least
approximately
0.700, at least approximately 0.750, at least approximately 0.800, at least
approximately
0.850, at least approximately 0.900, at least approximately 0.910, at least
approximately
0.920, at least approximately 0.930, at least approximately 0.940, at least
approximately
0.950, at least approximately 0.960, at least approximately 0.970, at least
approximately
0.980, at least approximately 0.990, or at least approximately 0.995.
[0054] "Bead" and "particle" are used herein interchangeably and refer to a
substantially
spherical solid support. One example of a bead or particle is a microparticle.
Microparticles
that can be used herein can be any type known in the art. For example, the
bead or particle
can be a magnetic bead or magnetic particle. Magnetic beads/particles may be
ferromagnetic, ferrimagnetic, paramagnetic, superparamagnetic or ferrofluidic.
Exemplary
ferromagnetic materials include Fe, Co, Ni, Gd, Dy, Cr02, MnAs, MnBi, Eu0, and
NiO/Fe.
Examples of ferrimagnetic materials include NiFe204, CoFe204, Fe304 (or
Fe0"Fe203).
Beads can have a solid core portion that is magnetic and is surrounded by one
or more non-
magnetic layers. Alternately, the magnetic portion can be a layer around a non-
magnetic
core. The microparticles can be of any size that would work in the methods
described herein,
e.g., from about 0.75 to about 5 nm, or from about 1 to about 5 nm, or from
about 1 to about
3 nm.
[0055] "Binding protein" is used herein to refer to a monomeric or
multimeric protein that
binds to and forms a complex with a binding partner, such as, for example, a
polypeptide, an
antigen, a chemical compound or other molecule, or a substrate of any kind. A
binding
protein specifically binds a binding partner. Binding proteins include
antibodies, as well as
antigen-binding fragments thereof and other various forms and derivatives
thereof as are
known in the art and described herein below, and other molecules comprising
one or more
antigen-binding domains that bind to an antigen molecule or a particular site
(epitope) on the
antigen molecule. Accordingly, a binding protein includes, but is not limited
to, an antibody
a tetrameric immunoglobulin, an IgG molecule, an IgG1 molecule, a monoclonal
antibody, a
12

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
chimeric antibody, a CDR-grafted antibody, a humanized antibody, an affinity
matured
antibody, and fragments of any such antibodies that retain the ability to bind
to an antigen.
[0056] "Bispecific antibody" is used herein to refer to a full-length
antibody that is generated
by quadroma technology (see Milstein etal., Nature, 305(5934): 537-540
(1983)), by
chemical conjugation of two different monoclonal antibodies (see, Staerz et
al., Nature,
314(6012): 628-631 (1985)), or by knob-into-hole or similar approaches, which
introduce
mutations in the Fc region (see Holliger etal., Proc. Natl. Acad. Sci. USA,
90(14): 6444-6448
(1993)), resulting in multiple different immunoglobulin species of which only
one is the
functional bispecific antibody. A bispecific antibody binds one antigen (or
epitope) on one of
its two binding arms (one pair of HC/LC), and binds a different antigen (or
epitope) on its
second arm (a different pair of HC/LC). By this definition, a bispecific
antibody has two
distinct antigen-binding arms (in both specificity and CDR sequences), and is
monovalent for
each antigen to which it binds to.
[0057] "CDR" is used herein to refer to the "complementarity determining
region" within an
antibody variable sequence. There are three CDRs in each of the variable
regions of the
heavy chain and the light chain. Proceeding from the N-terminus of a heavy or
light chain,
these regions are denoted "CDR1", "CDR2", and "CDR3", for each of the variable
regions.
The term "CDR set" as used herein refers to a group of three CDRs that occur
in a single
variable region that binds the antigen. An antigen-binding site, therefore,
may include six
CDRs, comprising the CDR set from each of a heavy and a light chain variable
region. A
polypeptide comprising a single CDR, (e.g., a CDR1, CDR2, or CDR3) may be
referred to as
a "molecular recognition unit." Crystallographic analyses of antigen-antibody
complexes
have demonstrated that the amino acid residues of CDRs form extensive contact
with bound
antigen, wherein the most extensive antigen contact is with the heavy chain
CDR3. Thus, the
molecular recognition units may be primarily responsible for the specificity
of an antigen-
binding site. In general, the CDR residues are directly and most substantially
involved in
influencing antigen binding.
[0058] The exact boundaries of these CDRs have been defined differently
according to
different systems. The system described by Kabat (Kabat et al., Sequences of
Proteins of
Immunological Interest (National Institutes of Health, Bethesda, Md. (1987)
and (1991)) not
13

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
only provides an unambiguous residue numbering system applicable to any
variable region of
an antibody, but also provides precise residue boundaries defining the three
CDRs. These
CDRs may be referred to as "Kabat CDRs". Chothia and coworkers (Chothia and
Lesk, J.
Mol. Biol., 196: 901-917 (1987); and Chothia et al., Nature, 342: 877-883
(1989)) found that
certain sub-portions within Kabat CDRs adopt nearly identical peptide backbone
conformations, despite having great diversity at the level of amino acid
sequence. These sub-
portions were designated as "Ll", "L2", and "L3", or "Hl", "H2", and "H3",
where the "L"
and the "H" designate the light chain and the heavy chain regions,
respectively. These
regions may be referred to as "Chothia CDRs", which have boundaries that
overlap with
Kabat CDRs. Other boundaries defining CDRs overlapping with the Kabat CDRs
have been
described by Padlan, FASEB J., 9: 133-139 (1995), and MacCallum, I Mol. Biol.,
262(5):
732-745 (1996). Still other CDR boundary definitions may not strictly follow
one of the
herein systems, but will nonetheless overlap with the Kabat CDRs, although
they may be
shortened or lengthened in light of prediction or experimental findings that
particular
residues or groups of residues or even entire CDRs do not significantly impact
antigen
binding. The methods used herein may utilize CDRs defined according to any of
these
systems, although certain embodiments use Kabat- or Chothia-defined CDRs.
[0059] "Component," "components," or "at least one component," refer
generally to a capture
antibody, a detection or conjugate a calibrator, a control, a sensitivity
panel, a container, a
buffer, a diluent, a salt, an enzyme, a co-factor for an enzyme, a detection
reagent, a
pretreatment reagent/solution, a substrate (e.g., as a solution), a stop
solution, and the like
that can be included in a kit for assay of a test sample, such as a patient
urine, whole blood,
serum or plasma sample, in accordance with the methods described herein and
other methods
known in the art. Some components can be in solution or lyophilized for
reconstitution for
use in an assay.
[0060] "Correlated to" as used herein refers to compared to.
[0061] "CT scan" as used herein refers to a computerized tomography (CT) scan.
A CT scan
combines a series of X-ray images taken from different angles and uses
computer processing
to create cross-sectional images, or slices, of the bones, blood vessels and
soft tissues inside
your body. The CT scan may use X-ray CT, positron emission tomography (PET),
single-
14

CA 03068041 2019-12-19
WO 2019/010131
PCT/US2018/040612
photon emission computed tomography (SPECT), computed axial tomography (CAT
scan),
or computer aided tomography. The CT scan may be a conventional CT scan or a
spiral/helical CT scan. In a conventional CT scan, the scan is taken slice by
slice and after
each slice the scan stops and moves down to the next slice, e.g., from the top
of the abdomen
down to the pelvis. The conventional CT scan requires patients to hold their
breath to avoid
movement artefact. The spiral/helical CT scan is a continuous scan which is
taken in a spiral
fashion and is a much quicker process where the scanned images are contiguous.
[0062]
"Determined by an assay" is used herein to refer to the determination of a
reference
level by any appropriate assay. The determination of a reference level may, in
some
embodiments, be achieved by an assay of the same type as the assay that is to
be applied to
the sample from the subject (for example, by an immunoassay, clinical
chemistry assay, a
single molecule detection assay, protein immunoprecipitation,
immunoelectrophoresis,
chemical analysis, SDS-PAGE and Western blot analysis, or protein
immunostaining,
electrophoresis analysis, a protein assay, a competitive binding assay, a
functional protein
assay, or chromatography or spectrometry methods, such as high-performance
liquid
chromatography (HPLC) or liquid chromatography¨mass spectrometry (LC/MS)). The
determination of a reference level may, in some embodiments, be achieved by an
assay of the
same type and under the same assay conditions as the assay that is to be
applied to the sample
from the subject. As noted herein, this disclosure provides exemplary
reference levels (e.g.,
calculated by comparing reference levels at different time points). It is well
within the
ordinary skill of one in the art to adapt the disclosure herein for other
assays to obtain assay-
specific reference levels for those other assays based on the description
provided by this
disclosure. For example, a set of training samples comprising samples obtained
from human
subjects known to have sustained an injury to the head (and more particularly,
samples
obtained from human subjects known to have sustained a (i) mild TB!; and/or
(ii) moderate,
severe, or moderate to severe TBI and samples obtained from human subjects
known not to
have sustained an injury to the head may be used to obtain assay-specific
reference levels. It
will be understood that a reference level "determined by an assay" and having
a recited level
of "sensitivity" and/or "specificity" is used herein to refer to a reference
level which has been
determined to provide a method of the recited sensitivity and/or specificity
when said

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
reference level is adopted in the methods of the invention. It is well within
the ordinary skill
of one in the art to determine the sensitivity and specificity associated with
a given reference
level in the methods of the invention, for example by repeated statistical
analysis of assay
data using a plurality of different possible reference levels.
[0063] Practically, when discriminating between a subject as having a
traumatic brain injury
or not having a traumatic brain injury or a subject as having a a mild versus
a moderate,
severe, or moderate to severe traumatic brain injury, the skilled person will
balance the effect
of raising a cutoff on sensitivity and specificity. Raising or lowering a
cutoff will have a
well-defined and predictable impact on sensitivity and specificity, and other
standard
statistical measures. It is well known that raising a cutoff will improve
specificity but is
likely to worsen sensitivity (proportion of those with disease who test
positive). In contrast,
lowering a cutoff will improve sensitivity but will worsen specificity
(proportion of those
without disease who test negative). The ramifications for detecting traumatic
brain injury or
determining a mild versus moderate, severe, or moderate to severe traumatic
brain injury will
be readily apparent to those skilled in the art. In discriminating whether a
subject has or does
not have a traumatic brain injury or a mild versus a moderate, severe, or
moderate to severe
traumatic brain injury, the higher the cutoff, specificity improves as more
true negatives (i.e.,
subjects not having a traumatic brain injury, not having a mild traumatic
brain injury, not
have a moderate traumatic brain injury, not having a severe traumatic brain
injury or not
having a moderate to severe traumatic brain injury) are distinguished from
those having a
traumatic brain injury, a mild traumatic brain injury, a moderate traumatic
brain injury, a
severe traumatic brain injury or a moderate to severe traumatic brain injury.
But at the same
time, raising the cutoff decreases the number of cases identified as positive
overall, as well as
the number of true positives, so the sensitivity must decrease. Conversely,
the lower the
cutoff, sensitivity improves as more true positives (i.e., subjects having a
traumatic brain
injury, having a mild traumatic brain injury, having a moderate traumatic
brain injury, having
a severe traumatic brain injury or having a moderate to severe traumatic brain
injury) are
distinguished from those who do not have a traumatic brain injury, a mild
traumatic brain
injury, a moderate traumatic brain injury, a severe traumatic brain injury or
a moderate to
severe traumatic brain injury. But at the same time, lowering the cutoff
increases the number
16

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
of cases identified as positive overall, as well as the number of false
positives, so the
specificity must decrease.
[0064] Generally, a high sensitivity value helps one of skill rule out
disease or condition (such
as a traumatic brain injury, mild traumatic brain injury, moderate traumatic
brain injury,
severe traumatic brain injury or moderate to severe traumatic brain injury),
and a high
specificity value helps one of skill rule in disease or condition. Whether one
of skill desires
to rule out or rule in disease depends on what the consequences are for the
patient for each
type of error. Accordingly, one cannot know or predict the precise balancing
employed to
derive a test cutoff without full disclosure of the underlying information on
how the value
was selected. The balancing of sensitivity against specificity and other
factors will differ on
a case-by-case basis. This is why it is sometimes preferable to provide
alternate cutoff (e.g.,
reference) values so a physician or practitioner can choose.
[0065] "Derivative" of an antibody as used herein may refer to an antibody
having one or
more modifications to its amino acid sequence when compared to a genuine or
parent
antibody and exhibit a modified domain structure. The derivative may still be
able to adopt
the typical domain configuration found in native antibodies, as well as an
amino acid
sequence, which is able to bind to targets (antigens) with specificity.
Typical examples of
antibody derivatives are antibodies coupled to other polypeptides, rearranged
antibody
domains, or fragments of antibodies. The derivative may also comprise at least
one further
compound, e.g., a protein domain, said protein domain being linked by covalent
or non-
covalent bonds. The linkage can be based on genetic fusion according to the
methods known
in the art. The additional domain present in the fusion protein comprising the
antibody may
preferably be linked by a flexible linker, advantageously a peptide linker,
wherein said
peptide linker comprises plural, hydrophilic, peptide-bonded amino acids of a
length
sufficient to span the distance between the C-teiminal end of the further
protein domain and
the N-terminal end of the antibody or vice versa. The antibody may be linked
to an effector
molecule having a conformation suitable for biological activity or selective
binding to a solid
support, a biologically active substance (e.g., a cytokine or growth hormone),
a chemical
agent, a peptide, a protein, or a drug, for example.
17

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0066] "Drugs of abuse" is used herein to refer to one or more additive
substances (such as a
drug) taken for non-medical reasons (such as for, example, recreational and/or
mind-altering
effects). Excessive overindulgence, use or dependence of such drugs of abuse
is often
referred to as "substance abuse". Examples of drugs of abuse include alcohol,
barbiturates,
benzodiazepines, cannabis, cocaine, hallucinogens (such as ketamine, mescaline
(peyote),
PCP, psilocybin, DMT and/or LSD), methaqualone, opioids, amphetamines
(including
methamphetamines), anabolic steroids, inhalants (namely, substances which
contain volatile
substances that contain psychoactive properties such as, for example,
nitrites, spray paints,
cleaning fluids, markers, glues, etc.) and combinations thereof
[0067] "Dual-specific antibody" is used herein to refer to a full-length
antibody that can bind
two different antigens (or epitopes) in each of its two binding arms (a pair
of HC/LC) (see
PCT publication WO 02/02773). Accordingly, a dual-specific binding protein has
two
identical antigen binding arms, with identical specificity and identical CDR
sequences, and is
bivalent for each antigen to which it binds.
[0068] "Dual variable domain" is used herein to refer to two or more
antigen binding sites on
a binding protein, which may be divalent (two antigen binding sites),
tetravalent (four
antigen binding sites), or multivalent binding proteins. DVDs may be
monospecific, i.e.,
capable of binding one antigen (or one specific epitope), or multispecific,
i.e., capable of
binding two or more antigens (i.e., two or more epitopes of the same target
antigen molecule
or two or more epitopes of different target antigens). A preferred DVD binding
protein
comprises two heavy chain DVD polypeptides and two light chain DVD
polypeptides and is
referred to as a "DVD immunoglobulin" or "DVD-Ig." Such a DVD-Ig binding
protein is
thus tetrameric and reminiscent of an IgG molecule, but provides more antigen
binding sites
than an IgG molecule. Thus, each half of a tetrameric DVD-Ig molecule is
reminiscent of
one half of an IgG molecule and comprises a heavy chain DVD polypeptide and a
light chain
DVD polypeptide, but unlike a pair of heavy and light chains of an IgG
molecule that
provides a single antigen binding domain, a pair of heavy and light chains of
a DVD-Ig
provide two or more antigen binding sites.
[0069] Each antigen binding site of a DVD-Ig binding protein may be derived
from a donor
("parental") monoclonal antibody and thus comprises a heavy chain variable
domain (VH)
18

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
and a light chain variable domain (VL) with a total of six CDRs involved in
antigen binding
per antigen binding site. Accordingly, a DVD-Ig binding protein that binds two
different
epitopes (i.e., two different epitopes of two different antigen molecules or
two different
epitopes of the same antigen molecule) comprises an antigen binding site
derived from a first
parental monoclonal antibody and an antigen binding site of a second parental
monoclonal
antibody.
[0070] A description of the design, expression, and characterization of DVD-
Ig binding
molecules is provided in PCT Publication No. WO 2007/024715, U.S. Patent No.
7,612,181,
and Wu et al., Nature Biotech., 25: 1290-1297 (2007). A preferred example of
such DVD-Ig
molecules comprises a heavy chain that comprises the structural formula VD1-
(Xl)n-VD2-
C-(X2)n, wherein VD1 is a first heavy chain variable domain, VD2 is a second
heavy chain
variable domain, C is a heavy chain constant domain, X1 is a linker with the
proviso that it is
not CH1, X2 is an Fc region, and n is 0 or 1, but preferably 1; and a light
chain that
comprises the structural formula VD1-(X1)n-VD2-C-(X2)n, wherein VD1 is a first
light
chain variable domain, VD2 is a second light chain variable domain, C is a
light chain
constant domain, X1 is a linker with the proviso that it is not CH1, and X2
does not comprise
an Fc region; and n is 0 or 1, but preferably 1. Such a DVD-Ig may comprise
two such heavy
chains and two such light chains, wherein each chain comprises variable
domains linked in
tandem without an intervening constant region between variable regions,
wherein a heavy
chain and a light chain associate to form tandem functional antigen binding
sites, and a pair
of heavy and light chains may associate with another pair of heavy and light
chains to form a
tetrameric binding protein with four functional antigen binding sites. In
another example, a
DVD-Ig molecule may comprise heavy and light chains that each comprise three
variable
domains (VD1, VD2, VD3) linked in tandem without an intervening constant
region between
variable domains, wherein a pair of heavy and light chains may associate to
form three
antigen binding sites, and wherein a pair of heavy and light chains may
associate with
another pair of heavy and light chains to fol in a tetrameric binding
protein with six antigen
binding sites.
[0071] In a preferred embodiment, a DVD-Ig binding protein not only binds
the same target
molecules bound by its parental monoclonal antibodies, but also possesses one
or more
119

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
desirable properties of one or more of its parental monoclonal antibodies.
Preferably, such
an additional property is an antibody parameter of one or more of the parental
monoclonal
antibodies. Antibody parameters that may be contributed to a DVD-Ig binding
protein from
one or more of its parental monoclonal antibodies include, but are not limited
to, antigen
specificity, antigen affinity, potency, biological function, epitope
recognition, protein
stability, protein solubility, production efficiency, immunogenicity,
pharmacokinetics,
bioavailability, tissue cross reactivity, and orthologous antigen binding.
[0072] A DVD-Ig binding protein binds at least one epitope of UCH-Li. Non-
limiting
examples of a DVD-Ig binding protein include a DVD-Ig binding protein that
binds one or
more epitopes of UCH-L1, a DVD-Ig binding protein that binds an epitope of a
human UCH-
Li and an epitope of UCH-L1 of another species (for example, mouse), and a DVD-
Ig
binding protein that binds an epitope of a human UCH-L1 and an epitope of
another target
molecule.
[0073] "Dynamic range" as used herein refers to a range over which an assay
readout is
proportional to the amount of target molecule or analyte in the sample being
analyzed.
100741 "Epitope," or "epitopes," or "epitopes of interest" refer to a
site(s) on any molecule
that is recognized and can bind to a complementary site(s) on its specific
binding partner.
The molecule and specific binding partner are part of a specific binding pair.
For example,
an epitope can be on a polypeptide, a protein, a hapten, a carbohydrate
antigen (such as, but
not limited to, glycolipids, glycoproteins or lipopolysaccharides), or a
polysaccharide. Its
specific binding partner can be, but is not limited to, an antibody.
[0075] "Expanded window of detection" as used herein refers to the fact
that the described
and/or claimed improved methods can be carried out in or under a variety of
clinical
scenarios when compared to other UCH-L1 assays. For example, the methods of
the present
disclosure can be carried out on any subject without regard to factors
selected from the group
consisting of the subject's clinical condition (e.g., whether or not there are
comorbid
conditions in addition to the reason for checking on UCH-L1), the subject's
laboratory values
(e.g., laboratory values other than UCH-L1 levels, including but not limited
to values on
standard laboratory tests that are run to assess a patient's overall health,
and values on more
particularized tests that are run when a subject is suspected of having been
in an accident or

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
exposed to some sort of trauma including but not limited to those that may
result in head
injury), the subject's classification as suffering from mild, moderate,
severe, moderate or
severe or moderate to severe TBI, the subject's exhibition (e.g.,
demonstration or possession)
of low or high levels of UCH-L1, and the timing of any event (e.g., relative
to testing) where
the subject may have sustained an injury to the head. Additionally, by way of
a further
example, the methods of the present disclosure differ from other methods known
in the prior
art which may or require dilution, or alternately, may lack one or more of the
benefits of the
improved assays as described herein (namely, measure up to 25,000 pg/mL,
dynamic range
of 5 log, assay linearity over the dynamic range, measure of UCH-L1 in a
volume less than
20 microliters of sample, expanded window of detection, etc.).
[0076] "Fragment antigen-binding fragment" or "Fab fragment" as used herein
refers to a
fragment of an antibody that binds to antigens and that contains one antigen-
binding site, one
complete light chain, and part of one heavy chain. Fab is a monovalent
fragment consisting
of the VL, VH, CL and CH1 domains. Fab is composed of one constant and one
variable
domain of each of the heavy and the light chain. The variable domain contains
the paratope
(the antigen-binding site), comprising a set of complementarity determining
regions, at the
amino terminal end of the monomer. Each arm of the Y thus binds an epitope on
the antigen.
Fab fragments can be generated such as has been described in the art, e.g.,
using the enzyme
papain, which can be used to cleave an immunoglobulin monomer into two Fab
fragments
and an Fc fragment, or can be produced by recombinant means.
[0077] "F(ab')2 fragment" as used herein refers to antibodies generated by
pepsin digestion of
whole IgG antibodies to remove most of the Fe region while leaving intact some
of the hinge
region. F(ab')2 fragments have two antigen-binding F(ab) portions linked
together by
disulfide bonds, and therefore are divalent with a molecular weight of about
110 kDa.
Divalent antibody fragments (F(ab')2 fragments) are smaller than whole IgG
molecules and
enable a better penetration into tissue thus facilitating better antigen
recognition in
immunohistochemistry. The use of F(a131)2 fragments also avoids unspecific
binding to Fe
receptor on live cells or to Protein A/G. F(ab')2 fragments can both bind and
precipitate
antigens.
21

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0078] "Framework" (FR) or "Framework sequence" as used herein may mean the
remaining
sequences of a variable region minus the CDRs. Because the exact definition of
a CDR
sequence can be determined by different systems (for example, see above), the
meaning of a
framework sequence is subject to correspondingly different interpretations.
The six CDRs
(CDR-L1, -L2, and -L3 of light chain and CDR-H1, -H2, and -H3 of heavy chain)
also divide
the framework regions on the light chain and the heavy chain into four sub-
regions (FR1,
FR2, FR3, and FR4) on each chain, in which CDR1 is positioned between FR1 and
IFR2,
CDR2 between FR2 and FR3, and CDR3 between FR3 and FR4. Without specifying the
particular sub-regions as FR1, FR2, FR3, or FR4, a framework region, as
referred by others,
represents the combined FRs within the variable region of a single, naturally
occurring
immunoglobulin chain. As used herein, a FR represents one of the four sub-
regions, and FRs
represents two or more of the four sub-regions constituting a framework
region.
[0079] Human heavy chain and light chain FR sequences are known in the art
that can be
used as heavy chain and light chain "acceptor" framework sequences (or simply,
"acceptor"
sequences) to humanize a non-human antibody using techniques known in the art.
In one
embodiment, human heavy chain and light chain acceptor sequences are selected
from the
framework sequences listed in publicly available databases such as V-base
(hypertext
transfer protocol://vbase.mrc-cpe.cam.ac.uld; phone number: +49 - 531 - 6181
687) or in the
international ImMunoGeneTicse (IMGTO) information system (hypertext transfer
protocol://imgt.cines.fr/texts/IMGTrepertoire/LocusGenes/; phone number: +33
(0)4 34 35
99 65).
[0080] "Functional antigen binding site" as used herein may mean a site on
a binding protein
(e.g., an antibody) that is capable of binding a target antigen. The antigen
binding affinity of
the antigen binding site may not be as strong as the parent binding protein,
e.g., parent
antibody, from which the antigen binding site is derived, but the ability to
bind antigen must
be measurable using any one of a variety of methods known for evaluating
protein, e.g.,
antibody, binding to an antigen. Moreover, the antigen binding affinity of
each of the antigen
binding sites of a multivalent protein, e.g., multivalent antibody, herein
need not be
quantitatively the same.
22

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0081] "Glasgow Coma Scale" or "GCS" as used herein refers to a 15 point
scale for
estimating and categorizing the outcomes of brain injury on the basis of
overall social
capability or dependence on others. The test measures the motor response,
verbal response
and eye opening response with these values: I. Motor Response (6 ¨ Obeys
commands fully;
¨ Localizes to noxious stimuli; 4 ¨ Withdraws from noxious stimuli; 3 ¨
Abnormal flexion,
i.e., decorticate posturing; 2 ¨ Extensor response, i.e., decerebrate
posturing; and 1 ¨ No
response); II. Verbal Response (5 ¨ Alert and Oriented; 4 ¨ Confused, yet
coherent, speech; 3
¨ Inappropriate words and jumbled phrases consisting of words; 2 ¨
Incomprehensible
sounds; and 1 ¨ No sounds); and III. Eye Opening (4 ¨ Spontaneous eye opening;
3 ¨ Eyes
open to speech; 2 ¨ Eyes open to pain; and 1 ¨ No eye opening). The final
score is
determined by adding the values of Id-ID-III. The final score can be
categorized into four
possible levels for survival, with a lower number indicating a more severe
injury and a poorer
prognosis: Mild (13-15); Moderate Disability (9-12) (Loss of consciousness
greater than 30
minutes; Physical or cognitive impairments which may or may resolve: and
Benefit from
Rehabilitation); Severe Disability (3-8) (Coma: unconscious state. No
meaningful response,
no voluntary activities); and Vegetative State (Less Than 3) (Sleep wake
cycles; Arousal, but
no interaction with environment; No localized response to pain). Moderate
brain injury is
defined as a brain injury resulting in a loss of consciousness from 20 minutes
to 6 hours and a
Glasgow Coma Scale of 9 to 12. Severe brain injury is defined as a brain
injury resulting in a
loss of consciousness of greater than 6 hours and a Glasgow Coma Scale of 3 to
8.
[0082] "Glasgow Outcome Scale" as used herein refers to a global scale for
functional
outcome that rates patient status into one of five categories: Dead,
Vegetative State, Severe
Disability, Moderate Disability or Good Recovery.
[0083] "Extended Glasgow Outcome Scale" or "GOSE" as used interchangeably
herein
provides more detailed categorization into eight categories by subdividing the
categories of
severe disability, moderate disability and good recovery into a lower and
upper category as
shown in Table 1.
Table 1
1 Death
Condition of unawareness with only reflex
2 Vegetative state VX
responses but with periods of spontaneous
23

CA 03068041 2019-12-19
WO 2019/010131
PCT/US2018/040612
eye opening
3 Lower severe disability SD -
Patient who is dependent for daily support
for mental or physical disability, usually a
combination of both. If the patient can be left
4 Upper severe disability SD + alone for more than 8 hours at home
it is
upper level of SD, if not then it is low level
of SD.
Patients have some disability such as
Lower moderate disability MD - aphasia, hemiparesis or epilepsy and/or
deficits of memory or personality but are able
to look after themselves. They are
independent at home but dependent outside.
6 Upper moderate disability MD
If they are able to return to work even with
special arrangement it is upper level of MD,
if not then it is low level of MD.
Resumption of normal life with the capacity
to work even if pre-injury status has not been
7 Lower good recovery GR -
achieved. Some patients have minor
neurological or psychological deficits. If
these deficits are not disabling then it is
8 Upper good recovery GR + upper level of GR, if disabling then it
is
lower level of GR.
[0084]
"Humanized antibody" is used herein to describe an antibody that comprises
heavy
and light chain variable region sequences from a non-human species (e.g., a
mouse) but in
which at least a portion of the VH and/or VL sequence has been altered to be
more "human-
like," i.e., more similar to human germline variable sequences. A "humanized
antibody" is
an antibody or a variant, derivative, analog, or fragment thereof, which
immunospecifically
binds to an antigen of interest and which comprises a framework (FR) region
having
substantially the amino acid sequence of a human antibody and a complementary
determining region (CDR) having substantially the amino acid sequence of a non-
human
antibody. As used herein, the term "substantially" in the context of a CDR
refers to a CDR
having an amino acid sequence at least 80%, at least 85%, at least 90%, at
least 95%, at least
98 A), or at least 99% identical to the amino acid sequence of a non-human
antibody CDR. A
humanized antibody comprises substantially all of at least one, and typically
two, variable
domains (Fab, Fab', F(ab')2, FabC, Fv) in which all or substantially all of
the CDR regions
correspond to those of a non-human immunoglobulin (i.e., donor antibody) and
all or
24

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
substantially all of the framework regions are those of a human immunoglobulin
consensus
sequence. In an embodiment, a humanized antibody also comprises at least a
portion of an
immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
In some
embodiments, a humanized antibody contains the light chain as well as at least
the variable
domain of a heavy chain. The antibody also may include the CH1, hinge, CH2,
CH3, and
CH4 regions of the heavy chain. In some embodiments, a humanized antibody only
contains
a humanized light chain. In some embodiments, a humanized antibody only
contains a
humanized heavy chain. In specific embodiments, a humanized antibody only
contains a
humanized variable domain of a light chain and/or humanized heavy chain.
[0085] A humanized antibody can be selected from any class of
immunoglobulins, including
IgM, IgG, IgD, IgA, and IgE, and any isotype, including without limitation
IgGl, IgG2,
IgG3, and IgG4. A humanized antibody may comprise sequences from more than one
class
or isotype, and particular constant domains may be selected to optimize
desired effector
functions using techniques well-known in the art.
[0086] The framework regions and CDRs of a humanized antibody need not
correspond
precisely to the parental sequences, e.g., the donor antibody CDR or the
consensus
framework may be mutagenized by substitution, insertion, and/or deletion of at
least one
amino acid residue so that the CDR or framework residue at that site does not
correspond to
either the donor antibody or the consensus framework. In a preferred
embodiment, such
mutations, however, will not be extensive. Usually, at least 80%, preferably
at least 85%,
more preferably at least 90%, and most preferably at least 95% of the
humanized antibody
residues will correspond to those of the parental FR and CDR sequences. As
used herein, the
term "consensus framework" refers to the framework region in the consensus
immunoglobulin sequence. As used herein, the term "consensus immunoglobulin
sequence"
refers to the sequence formed from the most frequently occurring amino acids
(or
nucleotides) in a family of related immunoglobulin sequences (see, e.g.,
Winnaker, From
Genes to Clones (Verlagsgesellschaft, Weinheim, 1987)). A "consensus
immunoglobulin
sequence" may thus comprise a "consensus framework region(s)" and/or a
"consensus
CDR(s)". In a family of immunoglobulins, each position in the consensus
sequence is

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
occupied by the amino acid occurring most frequently at that position in the
family. If two
amino acids occur equally frequently, either can be included in the consensus
sequence.
[0087] "Identical" or "identity," as used herein in the context of two or
more polypeptide or
polynucleotide sequences, can mean that the sequences have a specified
percentage of
residues that are the same over a specified region. The percentage can be
calculated by
optimally aligning the two sequences, comparing the two sequences over the
specified
region, determining the number of positions at which the identical residue
occurs in both
sequences to yield the number of matched positions, dividing the number of
matched
positions by the total number of positions in the specified region, and
multiplying the result
by 100 to yield the percentage of sequence identity. In cases where the two
sequences are of
different lengths or the alignment produces one or more staggered ends and the
specified
region of comparison includes only a single sequence, the residues of the
single sequence are
included in the denominator but not the numerator of the calculation.
[0088] "Injury to the head" or "head injury" as used interchangeably
herein, refers to any
trauma to the scalp, skull, or brain. Such injuries may include only a minor
bump on the skull
or may be a serious brain injury. Such injuries include primary injuries to
the brain and/or
secondary injuries to the brain. Primary brain injuries occur during the
initial insult and
result from displacement of the physical structures of the brain. More
specifically, a primary
brain injury is the physical damage to parenchyma (tissue, vessels) that
occurs during the
traumatic event, resulting in shearing and compression of the surrounding
brain tissue.
Secondary brain injuries occur subsequent to the primary injury and may
involve an array of
cellular processes. More specifically, a secondary brain injury refers to the
changes that
evolve over a period of time (from hours to days) after the primary brain
injury. It includes
an entire cascade of cellular, chemical, tissue, or blood vessel changes in
the brain that
contribute to further destruction of brain tissue.
[0089] An injury to the head can be either closed or open (penetrating). A
closed head injury
refers to a trauma to the scalp, skull or brain where there is no penetration
of the skull by a
striking object. An open head injury refers a trauma to the scalp, skull or
brain where there is
penetration of the skull by a striking object. An injury to the head may be
caused by physical
shaking of a person, by blunt impact by an external mechanical or other force
that results in a
26

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
closed or open head trauma (e.g., vehicle accident such as with an automobile,
plane, train,
etc.; blow to the head such as with a baseball bat, or from a firealln), a
cerebral vascular
accident (e.g., stroke), one or more falls (e.g., as in sports or other
activities), explosions or
blasts (collectively, "blast injuries") and by other types of blunt force
trauma. Alternatively,
an injury to the head may be caused by the ingestion and/or exposure to a
chemical, toxin or
a combination of a chemical and toxin. Examples of such chemicals and/or
toxins include
fires, molds, asbestos, pesticides and insecticides, organic solvents, paints,
glues, gases (such
as carbon monoxide, hydrogen sulfide, and cyanide), organic metals (such as
methyl
mercury, tetraethyl lead and organic tin) and/or one or more drugs of abuse.
Alternatively,
an injury to the head may be caused as a result of a subject suffering from an
autoimmune
disease, a metabolic disorder, a brain tumor, one or more viruses, meningitis,
hydrocephalus,
hypoxia or any combinations thereof. In some cases, it is not possible to be
certain whether
any such event or injury has occurred or taken place. For example, there may
be no history
on a patient or subject, the subject may be unable to speak, the subject may
be aware of what
events they were exposed to, etc. Such circumstances are described herein as
the subject
"may have sustained an injury to the head." In certain embodiments herein, the
closed head
injury does not include and specifically excludes a cerebral vascular
accident, such as stroke.
[0090] "Isolated polynucleotide" as used herein may mean a polynucleotide
(e.g., of genomic,
cDNA, or synthetic origin, or a combination thereof) that, by virtue of its
origin, the isolated
polynucleotide is not associated with all or a portion of a polynucleotide
with which the
"isolated polynucleotide" is found in nature; is operably linked to a
polynucleotide that it is
not linked to in nature; or does not occur in nature as part of a larger
sequence.
[0091] "Label" and "detectable label" as used herein refer to a moiety
attached to an antibody
or an analyte to render the reaction between the antibody and the analyte
detectable, and the
antibody or analyte so labeled is referred to as "detectably labeled." A label
can produce a
signal that is detectable by visual or instrumental means. Various labels
include signal-
producing substances, such as chromagens, fluorescent compounds,
chemiluminescent
compounds, radioactive compounds, and the like. Representative examples of
labels include
moieties that produce light, e.g., acridinium compounds, and moieties that
produce
fluorescence, e.g., fluorescein. Other labels are described herein. In this
regard, the moiety,
27

itself, may not be detectable but may become detectable upon reaction with yet
another
moiety. Use of the term "detectably labeled" is intended to encompass such
labeling.
[0092] "Limit of Detection (LOD)" as used herein refers to the lowest
concentration of the
measured object (i.e., a quantity intended to be measured) that can be
detected at a specified
level of confidence. The level of confidence is typically 95%, with a 5%
likelihood of a false
negative measurement. The LoD term used herein is based on the definition from
Clinical
and Laboratory Standards Institute (CLSI) protocol EP I7-A2 ("Protocols for
Determination
of Limits of Detection and Limits of Quantitation; Approved Guideline - Second
Edition,"
EP17A2E, by James F. Pierson-Perry et al., Clinical and Laboratory Standards
Institute, June
1, 2012),
[0093] "Linear" means that there is less than or equal to about 20%, about
19%, about 18%,
about 17%, about 16%, about 15%, about 14%, about 13%, about 12%, about 11%,
about
10%, about 9%, or about 8% variation for or over an exemplary range or value
recited.
[0094] Any suitable detectable label as is known in the art can be used.
For example, the
detectable label can be a radioactive label (such as 3H, 14C, 32P, 33P, 35S,
90Y, 99Tc,
1111n, 1251, 1311, 177Lu, 166Ho, and 153Sm), an enzymatic label (such as
horseradish
peroxidase, alkaline peroxidase, glucose 6-phosphate dehydrogenase, and the
like), a
chemiluminescent label (such as acridinium esters, thioesters, or
sulfonamides; luminol,
isoluminol, phenanthridinium esters, and the like), a fluorescent label (such
as fluorescein
(e.g., 5-fluorescein, 6-carboxyfluorescein, 3'6-carboxyfluorescein, 5(6)-
carboxyfluorescein,
6-hexachloro-fluorescein, 6-tetrachlorofluorescein, fluorescein
isothiocyanate, and the like)),
rhodamine, phycobiliproteins, R-phycoerythrin, quantum dots (e.g., zinc
sulfide-capped
cadmium selenide), a thermometric label, or an immuno-polymerase chain
reaction label. An
introduction to labels, labeling procedures and detection of labels is found
in Polak and Van
Noorden, Introduction to Immunocytochemistry, 2nd ed., Springer Verlag, N.Y.
(1997), and
in Haugland, Handbook of Fluorescent Probes and Research Chemicals (1996),
which is a
combined handbook and catalogue published by Molecular Probes, Inc., Eugene,
Oregon. A
fluorescent label can be used in FPIA (see, e.g., U.S. Patent Nos. 5,593,896,
5,573,904,
5,496,925, 5,359,093, and 5,352,803).
An acridinium compound can be used as a detectable label in a homogeneous
28
Date Recue/Date Received 2023-06-29

chemiluminescent assay (see, e.g., Adamczyk et al., Bioorg. Med. Chem. Lett.
16: 1324-1328
(2006); Adamczyk et al., Bioorg. Med Chem. Lett. 4: 2313-2317 (2004); Adamczyk
et al.,
Biorg. Med. Chem. Lett. 14: 3917-3921 (2004); and Adamczyk etal., Org. Lett.
5: 3779-
3782 (2003)).
[0095] In one aspect, the acridinium compound is an acridinium-9-carboxamide.
Methods for
preparing acridinium 9-carboxamides are described in Mattingly, J. Biolumin.
Chemilumin.
6: 107-114 (1991); Adamczyk et al., Org. Chem. 63: 5636-5639 (1998); Adamczyk
et al.,
Tetrahedron 55: 10899-10914 (1999); Adamczyk etal., Org. Lett. 1: 779-781
(1999);
Adamczyk et al., Bioconjugate Chem. 11: 714-724 (2000); Mattingly et al., In
Luminescence
Biotechnology: Instruments and Applications; Dyke, K. V. Ed.; CRC Press: Boca
Raton, pp.
77-105 (2002); Adamczyk etal., Org. Lett. 5: 3779-3782 (2003); and U.S. Patent
Nos.
5,468,646, 5,543,524 and 5,783,699.
[0096] Another example of an acridinium compound is an acridinium-9-
carboxylate aryl
ester. An example of an acridinium-9-carboxylate aryl ester of formula His 10-
methyl-9-
(phenoxycarbonyl)acridinium fluorosulfonate (available from Cayman Chemical,
Ann Arbor,
MI). Methods for preparing acridinium 9-carboxylate aryl esters are described
in McCapra et
al., Photochem. Photobiol. 4: 1111-21 (1965); Razavi et al., Luminescence 15:
245-249
(2000); Razavi et al., Luminescence 15: 239-244 (2000); and U.S. Patent No.
5,241,070.
Such acridinium-9-carboxylate aryl esters are efficient chemiluminescent
indicators
for hydrogen peroxide produced in the oxidation of an analyte by at least one
oxidase in
terms of the intensity of the signal and/or the rapidity of the signal. The
course of the
chemiluminescent emission for the acridinium-9-carboxylate aryl ester is
completed rapidly,
i.e., in under 1 second, while the acridinium-9-carboxamide chemiluminescent
emission
extends over 2 seconds. Acridinium-9-carboxylate aryl ester, however, loses
its
chemiluminescent properties in the presence of protein. Therefore, its use
requires the
absence of protein during signal generation and detection. Methods for
separating or
removing proteins in the sample are well-known to those skilled in the art and
include, but
are not limited to, ultrafiltration, extraction, precipitation, dialysis,
chromatography, and/or
29
Date Recue/Date Received 2023-06-29

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
digestion (see, e.g., Wells, High Throughput Bioanalytical Sample Preparation.
Methods and
Automation Strategies, Elsevier (2003)). The amount of protein removed or
separated from
the test sample can be about 40%, about 45%, about 50%, about 55%, about 60%,
about
65%, about 70%, about 75%, about 80%, about 85%, about 90%, or about 95%.
Further
details regarding acridinium-9-carboxylate aryl ester and its use are set
forth in U.S. Patent
App. No. 11/697,835, filed April 9, 2007. Acridinium-9-carboxylate aryl esters
can be
dissolved in any suitable solvent, such as degassed anhydrous N,N-
dimethylformamide
(DMF) or aqueous sodium cholate.
100971 "Linking sequence" or "linking peptide sequence" refers to a natural
or artificial
polypeptide sequence that is connected to one or more polypeptide sequences of
interest
(e.g., full-length, fragments, etc.). The term "connected" refers to the
joining of the linking
sequence to the polypeptide sequence of interest. Such polypeptide sequences
are preferably
joined by one or more peptide bonds. Linking sequences can have a length of
from about 4
to about 50 amino acids. Preferably, the length of the linking sequence is
from about 6 to
about 30 amino acids. Natural linking sequences can be modified by amino acid
substitutions, additions, or deletions to create artificial linking sequences.
Linking sequences
can be used for many purposes, including in recombinant Fabs. Exemplary
linking
sequences include, but are not limited to: (i) Histidine (His) tags, such as a
6X His tag,
which has an amino acid sequence of HI-11-1HHH (SEQ ID NO:2), are useful as
linking
sequences to facilitate the isolation and purification of polypeptides and
antibodies of
interest; (ii) Enterokinase cleavage sites, like His tags, are used in the
isolation and
purification of proteins and antibodies of interest. Often, enterokinase
cleavage sites are used
together with His tags in the isolation and purification of proteins and
antibodies of interest.
Various enterokinase cleavage sites are known in the art. Examples of
enterokinase cleavage
sites include, but are not limited to, the amino acid sequence of DDDDK (SEQ
ID NO:3) and
derivatives thereof (e.g., ADDDDK (SEQ ID NO:4), etc.); (iii) Miscellaneous
sequences can
be used to link or connect the light and/or heavy chain variable regions of
single chain
variable region fragments. Examples of other linking sequences can be found in
Bird et al.,
Science 242: 423-426 (1988); Huston et al., PNAS USA 85: 5879-5883 (1988); and
McCafferty et al. , Nature 348: 552-554 (1990). Linking sequences also can be
modified for

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
additional functions, such as attachment of drugs or attachment to solid
supports. In the
context of the present disclosure, the monoclonal antibody, for example, can
contain a
linking sequence, such as a His tag, an enterokinase cleavage site, or both.
[0098] "Monoclonal antibody" as used herein refers to an antibody obtained
from a
population of substantially homogeneous antibodies, i.e., the individual
antibodies
comprising the population are identical except for possible naturally
occurring mutations that
may be present in minor amounts. Monoclonal antibodies are highly specific,
being directed
against a single antigen. Furthermore, in contrast to polyclonal antibody
preparations that
typically include different antibodies directed against different determinants
(epitopes), each
monoclonal antibody is directed against a single determinant on the antigen.
The monoclonal
antibodies herein specifically include "chimeric" antibodies in which a
portion of the heavy
and/or light chain is identical with or homologous to corresponding sequences
in antibodies
derived from a particular species or belonging to a particular antibody class
or subclass,
while the remainder of the chain(s) is identical with or homologous to
corresponding
sequences in antibodies derived from another species or belonging to another
antibody class
or subclass, as well as fragments of such antibodies, so long as they exhibit
the desired
biological.
[0099] "Magnetic resonance imaging" or "MRI" as used interchangeably herein
refers to
magnetic resonance imaging, which is a medical imaging technique used in
radiology to form
pictures of the anatomy and the physiological processes of the body in both
health and
disease. MRI is a form of medical imaging that measures the response of the
atomic nuclei
of body tissues to high-frequency radio waves when placed in a strong magnetic
field, and
that produces images of the internal organs. MRI scanners, which is based on
the science of
nuclear magnetic resonance (NMR), use strong magnetic fields, radio waves, and
field
gradients to generate images of the inside of the body.
[0100] "Multivalent binding protein" is used herein to refer to a binding
protein comprising
two or more antigen binding sites (also referred to herein as "antigen binding
domains"). A
multivalent binding protein is preferably engineered to have three or more
antigen binding
sites, and is generally not a naturally occurring antibody. The term
"multispecific binding
protein" refers to a binding protein that can bind two or more related or
unrelated targets,
31

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
including a binding protein capable of binding two or more different epitopes
of the same
target molecule.
[0101] "Negative predictive value" or "NPV" as used interchangeably herein
refers to the
probability that a subject has a negative outcome given that they have a
negative test result.
[0102] "Preanalytical processing conditions" as used interchangeably herein
refers to one or
more handling, temperature, chemical and/or environmental conditions that are
applied to or
conducted on a sample (such as a whole blood sample) in the period of time
between the time
the sample is obtained from the subject and the time point when an assay or
test is conducted
or performed on the sample. In one embodiment, a preanalytical processing
condition can be
processing a sample within no more than about eight hours after the sample is
obtained from
a subject. In another embodiment, a preanalytical processing condition can be
storing a
sample at room temperature in the period of time between the time the sample
is obtained
from the subject and the assay is performed. In another embodiment, a
preanalytical
processing condition can be storing a sample at 2-8 C in the period of time
between the time
the sample is obtained from the subject and the assay is performed. In another
embodiment,
a preanalytical processing condition can be not mixing the sample in the
period of time
between the time the sample is obtained from the subject and the time point
when the assay
or test is performed.
[0103] "Reference level" as used herein refers to an assay cutoff value
that is used to assess
diagnostic, prognostic, or therapeutic efficacy and that has been linked or is
associated with
various clinical parameters (e.g., presence of disease, stage of disease,
severity of disease,
progression, non-progression, or improvement of disease, etc.). This
disclosure provides
exemplary reference levels. However, it is well-known that reference levels
may vary
depending on the nature of the immunoassay (e.g., antibodies employed,
reaction conditions,
sample purity, etc.) and that assays can be compared and standardized. It
further is well
within the ordinary skill of one in the art to adapt the disclosure herein for
other
immunoassays to obtain immunoassay-specific reference levels for those other
immunoassays based on the description provided by this disclosure. Whereas the
precise
value of the reference level may vary between assays, the findings as
described herein should
be generally applicable and capable of being extrapolated to other assays.
32

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0104] "Point-of-care device" refers to a device used to provide medical
diagnostic testing at
or near the point-of-care (namely, outside of a laboratory), at the time and
place of patient
care (such as in a hospital, physician's office, urgent or other medical care
facility, a patient's
home, a nursing home and/or a long term care and/or hospice facility).
Examples of point-
of-care devices include those produced by Abbott Laboratories (Abbott Park,
IL) (e.g., i-
STAT and i-STAT Alinity, Universal Biosensors (Rowville, Australia) (see US
2006/0134713), Axis-Shield PoC AS (Oslo, Norway) and Clinical Lab Products
(Los
Angeles, USA).
[0105] "Positive predictive value" or "PPV" as used interchangeably herein
refers to the
probability that a subject has a positive outcome given that they have a
positive test result.
[0106] "Quality control reagents" in the context of immunoassays and kits
described herein,
include, but are not limited to, calibrators, controls, and sensitivity
panels. A "calibrator" or
"standard" typically is used (e.g., one or more, such as a plurality) in order
to establish
calibration (standard) curves for interpolation of the concentration of an
analyte, such as an
antibody or an analyte. Alternatively, a single calibrator, which is near a
reference level or
control level (e.g., "low," "medium," or "high" levels), can be used. Multiple
calibrators
(i.e., more than one calibrator or a varying amount of calibrator(s)) can be
used in
conjunction to comprise a "sensitivity panel."
[0107] A "receiver operating characteristic" curve or "ROC" curve refers to
a graphical plot
that illustrates the performance of a binary classifier system as its
discrimination threshold is
varied. For example, an ROC curve can be a plot of the true positive rate
against the false
positive rate for the different possible cutoff points of a diagnostic test.
It is created by
plotting the fraction of true positives out of the positives (TPR = true
positive rate) vs. the
fraction of false positives out of the negatives (FPR = false positive rate),
at various threshold
settings. TPR is also known as sensitivity, and FPR is one minus the
specificity or true
negative rate. The ROC curve demonstrates the tradeoff between sensitivity and
specificity
(any increase in sensitivity will be accompanied by a decrease in
specificity); the closer the
curve follows the left-hand border and then the top border of the ROC space,
the more
accurate the test; the closer the curve comes to the 45-degree diagonal of the
ROC space, the
33

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
less accurate the test; the slope of the tangent line at a cutoff point gives
the likelihood ratio
(LR) for that value of the test; and the area under the curve is a measure of
test accuracy.
[0108] "Recombinant antibody" and "recombinant antibodies" refer to
antibodies prepared by
one or more steps, including cloning nucleic acid sequences encoding all or a
part of one or
more monoclonal antibodies into an appropriate expression vector by
recombinant techniques
and subsequently expressing the antibody in an appropriate host cell. The
terms include, but
are not limited to, recombinantly produced monoclonal antibodies, chimeric
antibodies,
humanized antibodies (fully or partially humanized), multi-specific or multi-
valent structures
formed from antibody fragments, bifunctional antibodies, heteroconjugate Abs,
DVD-Iggs,
and other antibodies as described in (i) herein. (Dual-variable domain
immunoglobulins and
methods for making them are described in Wu, C., et al., Nature Biotechnology,
25:1290-
1297 (2007)). The term "bifunctional antibody," as used herein, refers to an
antibody that
comprises a first arm having a specificity for one antigenic site and a second
arm having a
specificity for a different antigenic site, i.e., the bifunctional antibodies
have a dual
specificity.
[0109] "Risk assessment," "risk classification," "risk identification," or
"risk stratification" of
subjects (e.g., patients) as used herein refers to the evaluation of factors
including
biomarkers, to predict the risk of occurrence of future events including
disease onset or
disease progression, so that treatment decisions regarding the subject may be
made on a more
informed basis.
[0110] "Sample," "test sample," "specimen," "biological sample", "sample
from a subject,"
and "patient sample" as used herein may be used interchangeable and may be a
sample of
blood, such as whole blood, tissue, urine, serum, plasma, amniotic fluid,
cerebrospinal fluid,
placental cells or tissue, endothelial cells, leukocytes, or monocytes. In
some embodiments,
the sample is a whole blood sample. In some embodiments, the sample is a serum
sample.
In yet other embodiments, the sample is a plasma sample. The sample can be
used directly as
obtained from a patient or can be pre-treated, such as by filtration,
distillation, extraction,
concentration, centrifugation, inactivation of interfering components,
addition of reagents,
and the like, to modify the character of the sample in some manner as
discussed herein or
otherwise as is known in the art.
34

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0111] A variety of cell types, tissue, or bodily fluid may be utilized to
obtain a sample. Such
cell types, tissues, and fluid may include sections of tissues such as biopsy
and autopsy
samples, frozen sections taken for histologic purposes, blood (such as whole
blood), plasma,
serum, red blood cells, platelets, interstitial fluid, cerebral spinal fluid,
etc. Cell types and
tissues may also include lymph fluid, cerebrospinal fluid, a fluid collected
by A tissue or cell
type may be provided by removing a sample of cells from a human and a non-
human animal,
but can also be accomplished by using previously isolated cells (e.g.,
isolated by another
person, at another time, and/or for another purpose). Archival tissues, such
as those having
treatment or outcome history, may also be used. Protein or nucleotide
isolation and/or
purification may not be necessary.
[0112] "Sensitivity" of an assay as used herein refers to the proportion of
subjects for whom
the outcome is positive that are correctly identified as positive (e.g.,
correctly identifying
those subjects with a disease or medical condition for which they are being
tested). For
example, this might include correctly identifying subjects as having a TBI
from those who do
not have a TBI, correctly identifying subjects having a moderate, severe, or
moderate to
severe TBI from those having a mild TBI, correctly identifying subjects as
having a mild TBI
from those having a moderate, severe, or moderate to severe TBI, correctly
identifying
subjects as having a moderate, severe, or moderate to severe TBI from those
having no TBI
or correctly identifying subjects as having a mild TBI from those having no
TBI, correctly
identifying subjects as likely to benefit from imaging or a head CT scan or a
MRI from those
who are not likely to benefit from a head imaging or a CT scan or MRI, etc.).
[0113] "Specificity" of an assay as used herein refers to the proportion of
subjects for whom
the outcome is negative that are correctly identified as negative (e.g.,
correctly identifying
those subjects who do not have a disease or medical condition for which they
are being
tested). For example, this might include correctly identifying subjects having
an TBI from
those who do not have a TBI, correctly identifying subjects not having a
moderate, severe, or
moderate to severe TBI from those having a mild TBI, correctly identifying
subjects as not
having a mild TBI from those having a moderate, severe, or moderate to severe
TBI or
correctly identifying subjects as not having any TBI, or correctly identifying
subjects as
having a mild TBI from those having no TBI, etc.).

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0114] "Series of calibrating compositions" refers to a plurality of
compositions comprising a
known concentration of UCH-LI, wherein each of the compositions differs from
the other
compositions in the series by the concentration of UCH-L1.
[0115] "Specific binding" or "specifically binding" as used herein may
refer to the interaction
of an antibody, a protein, or a peptide with a second chemical species,
wherein the interaction
is dependent upon the presence of a particular structure (e.g., an antigenic
detettninant or
epitope) on the chemical species; for example, an antibody recognizes and
binds to a specific
protein structure rather than to proteins generally. If an antibody is
specific for epitope "A",
the presence of a molecule containing epitope A (or free, unlabeled A), in a
reaction
containing labeled "A" and the antibody, will reduce the amount of labeled A
bound to the
antibody.
[0116] "Solid phase" or "solid support" as used interchangeably herein,
refers to any material
that can be used to attach and/or attract and immobilize (1) one or more
capture agents or
capture specific binding partners, or (2) one or more detection agents or
detection specific
binding partners. The solid phase can be chosen for its intrinsic ability to
attract and
immobilize a capture agent. Alternatively, the solid phase can have affixed
thereto a linking
agent that has the ability to attract and immobilize the (1) capture agent or
capture specific
binding partner, or (2) detection agent or detection specific binding partner.
For example, the
linking agent can include a charged substance that is oppositely charged with
respect to the
capture agent (e.g., capture specific binding partner) or detection agent
(e.g., detection
specific binding partner) itself or to a charged substance conjugated to the
(1) capture agent
or capture specific binding partner or (2) detection agent or detection
specific binding
partner. In general, the linking agent can be any binding partner (preferably
specific) that is
immobilized on (attached to) the solid phase and that has the ability to
immobilize the (1)
capture agent or capture specific binding partner, or (2) detection agent or
detection specific
binding partner through a binding reaction. The linking agent enables the
indirect binding of
the capture agent to a solid phase material before the performance of the
assay or during the
performance of the assay. For examples, the solid phase can be plastic,
derivatized plastic,
magnetic, or non-magnetic metal, glass or silicon, including, for example, a
test tube,
36

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
microtiter well, sheet, bead, microparticle, chip, and other configurations
known to those of
ordinary skill in the art.
[0117] "Specific binding partner" is a member of a specific binding pair. A
specific binding
pair comprises two different molecules, which specifically bind to each other
through
chemical or physical means. Therefore, in addition to antigen and antibody
specific binding
pairs of common immunoassays, other specific binding pairs can include biotin
and avidin
(or streptavidin), carbohydrates and lectins, complementary nucleotide
sequences, effector
and receptor molecules, cofactors and enzymes, enzymes and enzyme inhibitors,
and the like.
Furthermore, specific binding pairs can include members that are analogs of
the original
specific binding members, for example, an analyte-analog. Immunoreactive
specific binding
members include antigens, antigen fragments, and antibodies, including
monoclonal and
polyclonal antibodies as well as complexes and fragments thereof, whether
isolated or
recombinantly produced.
[0118] "Stability" as used herein refers to the change in concentration of
an analyte,
biomarker (e.g., such as UCH-Li) or target substance over time (such as in a
sample).
Stability can be determined by calculating the difference between a first test
value and one or
more subsequent test values (e.g., second test value, third test value, fourth
test value, fifth
test value, sixth test value, seventh test value, eighth test value, ninth
test value, tenth test
value, etc.). If the difference between the first test value and one or more
subsequent test
values is less than 10% (plus or minus), then the analyte, biomarker or target
substance in the
sample is considered to be stable. If the difference between the first test
value and one or
more subsequent test values is greater than 10% (plus or minus 10%), then the
analyte,
biomarker or target substance in the sample is not considered to be stable.
[0119] "Statistically significant" as used herein refers to the likelihood
that a relationship
between two or more variables is caused by something other than random chance.
Statistical
hypothesis testing is used to determine whether the result of a data set is
statistically
significant. In statistical hypothesis testing, a statistical significant
result is attained
whenever the observed p-value of a test statistic is less than the
significance level defined of
the study. Thep-value is the probability of obtaining results at least as
extreme as those
observed, given that the null hypothesis is true. Examples of statistical
hypothesis analysis
37

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
include Wilcoxon signed-rank test, t-test, Chi-Square or Fisher's exact test.
"Significant" as
used herein refers to a change that has not been determined to be
statistically significant (e.g.,
it may not have been subject to statistical hypothesis testing).
[0120] "Subject" and "patient" as used herein interchangeably refers to any
vertebrate,
including, but not limited to, a mammal (e.g., cow, pig, camel, llama, horse,
goat, rabbit,
sheep, hamsters, guinea pig, cat, dog, rat, and mouse, a non-human primate
(for example, a
monkey, such as a cynomolgous or rhesus monkey, chimpanzee, etc.) and a
human). In some
embodiments, the subject may be a human or a non-human. In some embodiments,
the
subject is a human. The subject or patient may be undergoing other forms of
treatment. In
some embodiments, when the subject is a human, the subject does not include
any humans
who have suffered a cerebrovascular accident (e.g., a stroke).
[0121] "Treat," "treating" or "treatment" are each used interchangeably
herein to describe
reversing, alleviating, or inhibiting the progress of a disease and/or injury,
or one or more
symptoms of such disease, to which such term applies. Depending on the
condition of the
subject, the term also refers to preventing a disease, and includes preventing
the onset of a
disease, or preventing the symptoms associated with a disease. A treatment may
be either
perfolined in an acute or chronic way. The term also refers to reducing the
severity of a
disease or symptoms associated with such disease prior to affliction with the
disease. Such
prevention or reduction of the severity of a disease prior to affliction
refers to administration
of a pharmaceutical composition to a subject that is not at the time of
administration afflicted
with the disease. "Preventing" also refers to preventing the recurrence of a
disease or of one
or more symptoms associated with such disease. "Treatment" and
"therapeutically," refer to
the act of treating, as "treating" is defined above. "Traumatic Brain Injury"
or "TBI" as used
interchangeably herein refers to a complex injury with a broad spectrum of
symptoms and
disabilities. TBI is most often an acute event similar to other injuries. TBI
can be classified
as "mild," "moderate," or "severe." The causes of TBI are diverse and include,
for example,
physical shaking by a person, a car accident, injuries from firearms, cerebral
vascular
accidents (e.g., strokes), falls, explosions or blasts and other types of
blunt force trauma.
Other causes of TBI include the ingestion and/or exposure to one or more
chemicals or toxins
(such as fires, molds, asbestos, pesticides and insecticides, organic
solvents, paints, glues,
38

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
gases (such as carbon monoxide, hydrogen sulfide, and cyanide), organic metals
(such as
methyl mercury, tetraethyl lead and organic tin), one or more drugs of abuse
or combinations
thereof). Alternatively, TBI can occur in subjects suffering from an
autoimmune disease, a
metabolic disorder, a brain tumor, hypoxia, one or more viruses, meningitis,
hydrocephalus
or combinations thereof. Young adults and the elderly are the age groups at
highest risk for
TBI. In certain embodiments herein, traumatic brain injury or TBI does not
include and
specifically excludes cerebral vascular accidents such as strokes.
[0122] "Mild TBI" as used herein refers to a brain injury where loss of
consciousness is brief
and usually a few seconds or minutes and/or confusion and disorientation is
shorter than 1
hour. Mild TBI is also referred to as a concussion, minor head trauma, minor
TBI, minor
brain injury, and minor head injury. While MRI and CT scans are often nolmal,
the
individual with mild TBI may have cognitive problems such as headache,
difficulty thinking,
memory problems, attention deficits, mood swings and frustration.
[0123] Mild TBI is the most prevalent TBI and is often missed at time of
initial injury.
Typically, a subject has a Glasgow Coma scale number of between 13-15 (such as
13-15 or
14-15). Fifteen percent (15%) of people with mild TBI have symptoms that last
3 months or
more. Mild TBI is defined as the result of the forceful motion of the head or
impact causing
a brief change in mental status (confusion, disorientation or loss of memory)
or loss of
consciousness for less than 30 minutes. Common symptoms of mild TBI include
fatigue,
headaches, visual disturbances, memory loss, poor attention/concentration,
sleep
disturbances, dizziness/loss of balance, irritability-emotional disturbances,
feelings of
depression, and seizures. Other symptoms associated with mild TBI include
nausea, loss of
smell, sensitivity to light and sounds, mood changes, getting lost or
confused, and/or
slowness in thinking.
[0124] "Moderate 1131" as used herein refers to a brain injury where loss
of consciousness
and/or confusion and disorientation is between 1 and 24 hours and the subject
has a Glasgow
Coma scale number of between 9-13 (such as 9-12 or 9-13). The individual with
moderate
TBI have abnormal brain imaging results. "Severe TBI" as used herein refers to
a brain
injury where loss of consciousness is more than 24 hours and memory loss after
the injury or
penetrating skull injury longer than 24 hours and the subject has a Glasgow
Coma scale
39

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
number between 3-8. The deficits range from impairment of higher level
cognitive functions
to comatose states. Survivors may have limited function of arms or legs,
abnormal speech or
language, loss of thinking ability or emotional problems. Individuals with
severe injuries can
be left in long-term unresponsive states. For many people with severe TBI,
long-term
rehabilitation is often necessary to maximize function and independence.
[0125] "Moderate to severe" TBI as used herein refers to a spectrum of
brain injury that
includes moderate to severe and thus encompasses moderate TBI alone, severe
TBI alone and
moderate to severe TBI combined. Subjects suffering from a moderate to severe
TBI can
have a Glasgow Coma scale number of between 3-13 (such as 3-12 or 3-13). For
example, in
some clinical situations, a subject may initially be diagnosed as having a
moderate TBI but
who, over the course of time (minutes, hours or days), progress to having a
severe TBI (such,
as for example, in situations when there is a brain bleed). Such subjects
would be examples
of patients that could be classified as "moderate to severe". Common symptoms
of moderate
to severe TBI include cognitive deficits including difficulties with
attention, concentration,
distractibility, memory, speed of processing, confusion, perseveration,
impulsiveness,
language processing, and/or "executive functions", not understanding the
spoken word
(receptive aphasia), difficulty speaking and being understood (expressive
aphasia), slurred
speech, speaking very fast or very slow, problems reading, problems writing,
difficulties with
interpretation of touch, temperature, movement, limb position and fine
discrimination, the
integration or patterning of sensory impressions into psychologically
meaningful data, partial
or total loss of vision, weakness of eye muscles and double vision (diplopia),
blurred vision,
problems judging distance, involuntary eye movements (nystagmus), intolerance
of light
(photophobia), hearing, such as decrease or loss of hearing, ringing in the
ears (tinnitus),
increased sensitivity to sounds, loss or diminished sense of smell (anosmia),
loss or
diminished sense of taste, the convulsions associated with epilepsy that can
be several types
and can involve disruption in consciousness, sensory perception, or motor
movements,
control of bowel and bladder, sleep disorders, loss of stamina, appetite
changes, regulation of
body temperature, menstrual difficulties, dependent behaviors, emotional
ability, lack of
motivation, irritability, aggression, depression, disinhibition, or
denial/lack of awareness.

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0126] "Ubiquitin carboxy-terminal hydrolase Li" or "UCH-Li" as used
interchangeably
herein refers to a deubiquitinating enzyme that is encoded by the UCH-Li gene
in humans,
and which can be produced (e.g., by recombinant means, in other species). UCH-
L1, also
known as ubiquitin carboxyl-terminal esterase Li and ubiquitin thiolesterase,
is a member of
a gene family whose products hydrolyze small C-terminal adducts of ubiquitin
to generate
the ubiquitin monomer.
[0127] "UCH-L1 status" can mean either the level or amount of UCH-L1 at a
point in time
(such as with a single measure of UCH-L1), the level or amount of UCH-L1
associated with
monitoring (such as with a repeat test on a subject to identify an increase or
decrease in
UCH-L1 amount), the level or amount of UCH-L1 associated with treatment for
traumatic
brain injury (whether a primary brain injury and/or a secondary brain injury)
or combinations
thereof
[0128] "Variant" is used herein to describe a peptide or polypeptide that
differs in amino acid
sequence by the insertion, deletion, or conservative substitution of amino
acids, but retain at
least one biological activity. Representative examples of "biological
activity" include the
ability to be bound by a specific antibody or to promote an immune response.
Variant is also
used herein to describe a protein with an amino acid sequence that is
substantially identical to
a referenced protein with an amino acid sequence that retains at least one
biological activity.
A conservative substitution of an amino acid, i.e., replacing an amino acid
with a different
amino acid of similar properties (e.g., hydrophilicity, degree, and
distribution of charged
regions) is recognized in the art as typically involving a minor change. These
minor changes
can be identified, in part, by considering the hydropathic index of amino
acids, as understood
in the art. Kyte etal., J. Mol. Biol. 157:105-132 (1982). The hydropathic
index of an amino
acid is based on a consideration of its hydrophobicity and charge. It is known
in the art that
amino acids of similar hydropathic indexes can be substituted and still retain
protein
function. In one aspect, amino acids having hydropathic indexes of 2 are
substituted. The
hydrophilicity of amino acids can also be used to reveal substitutions that
would result in
proteins retaining biological function. A consideration of the hydrophilicity
of amino acids
in the context of a peptide permits calculation of the greatest local average
hydrophilicity of
that peptide, a useful measure that has been reported to correlate well with
antigenicity and
41

immunogenicity. U.S. Patent No. 4,554,101.
Substitution of amino acids having similar hydrophilicity values can result in
peptides
retaining biological activity, for example immunogenicity, as is understood in
the art.
Substitutions may be performed with amino acids having hydrophilicity values
within 2 of
each other. Both the hydrophobicity index and the hydrophilicity value of
amino acids are
influenced by the particular side chain of that amino acid. Consistent with
that observation,
amino acid substitutions that are compatible with biological function are
understood to
depend on the relative similarity of the amino acids, and particularly the
side chains of those
amino acids, as revealed by the hydrophobicity, hydrophilicity, charge, size,
and other
properties. "Variant" also can be used to refer to an antigenically reactive
fragment of an
anti-UCH-L1 antibody that differs from the corresponding fragment of anti- UCH-
L1
antibody in amino acid sequence but is still antigenically reactive and can
compete with the
corresponding fragment of anti- UCH-L1 antibody for binding with UCH-Li.
"Variant" also
can be used to describe a polypeptide or a fragment thereof that has been
differentially
processed, such as by proteolysis, phosphorylation, or other post-
translational modification,
yet retains its antigen reactivity.
[0129] "Vector" is used herein to describe a nucleic acid molecule that can
transport another
nucleic acid to which it has been linked. One type of vector is a "plasmid",
which refers to a
circular double-stranded DNA loop into which additional DNA segments may be
ligated.
Another type of vector is a viral vector, wherein additional DNA segments may
be ligated
into the viral genome. Certain vectors can replicate autonomously in a host
cell into which
they are introduced (e.g., bacterial vectors having a bacterial origin of
replication and
episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian
vectors) can be
integrated into the genome of a host cell upon introduction into the host
cell, and thereby are
replicated along with the host genome. Moreover, certain vectors are capable
of directing the
expression of genes to which they are operatively linked. Such vectors are
referred to herein
as "recombinant expression vectors" (or simply, "expression vectors"). In
general,
expression vectors of utility in recombinant DNA techniques are often in the
form of
plasmids. "Plasmid" and "vector" may be used interchangeably as the plasmid is
the most
commonly used form of vector. However, other forms of expression vectors, such
as viral
42
Date Recue/Date Received 2023-06-29

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
vectors (e.g., replication defective retroviruses, adenoviruses and adeno-
associated viruses),
which serve equivalent functions, can be used. In this regard, RNA versions of
vectors
(including RNA viral vectors) may also find use in the context of the present
disclosure.
[0130] Unless otherwise defined herein, scientific and technical terms used
in connection with
the present disclosure shall have the meanings that are commonly understood by
those of
ordinary skill in the art. For example, any nomenclatures used in connection
with,
and techniques of, cell and tissue culture, molecular biology, immunology,
microbiology,
genetics and protein and nucleic acid chemistry and hybridization described
herein are
those that are well known and commonly used in the art. The meaning and scope
of the
terms should be clear; in the event, however of any latent ambiguity,
definitions provided
herein take precedent over any dictionary or extrinsic definition. Further,
unless otherwise
required by context, singular terms shall include pluralities and plural terms
shall include the
singular.
2. Preanalytical Processing Conditions Affecting Ubiquitin Carboxy-Terminal
Hydrolase
Li (UCH-L1) Levels in Samples and Methods of Measuring Ubiquitin Carboxy-
Terminal Hydrolase Li (UCH-L1) Levels
[0131] The present disclosure is directed to an improvement of a method of
measuring an
amount of UCH-L1 in a sample (e.g., blood sample (such as a whole blood, serum
or plasma
sample)) obtained from a subject. As described herein, certain preanalytical
processing
conditions or factors can influence UCH-L1 levels or concentrations, and in
some cases, lead
to increases in UCH-L1 levels or concentrations during or between the period
of time during
which the blood sample is being maintained or stored until a test or assay is
performed (such
as to detect and/or measure the amount of UCH-L1 in the sample). Such
increases in UCH-
Li levels or concentrations are erroneous (namely, an erroneous increase or
erroneously
high) and may not be an accurate reflection of a disease state (such as TBI)
and may lead to
false positive results. Thus, the present disclosure is also directed to
methods of avoiding or
preventing an erroneous increase or rise in UCH-L1 levels in a blood sample
obtained from a
subject prior to performing a test or assay to detect and/or measure UCH-Li
levels by
performing the methods described herein. Finally, the present disclosure is
also directed to
43

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
methods of stabilizing UCH-L1 levels in a blood sample to prevent or avoid an
increase or
rise in such levels during storage prior to performing a test or assay to
detect and/or measure
UCH-L1 levels in the blood sample.
101321 In one embodiment, a preanalytical processing condition or factor
discovered to
influence UCH-L1 levels or concentration is the processing time of the blood
sample,
namely, how quickly the blood sample is processed after it is obtained from a
subject
(namely, prior to testing or perfoiming an assay). In some embodiments, it is
advantageous
to process a blood sample (e.g., a whole blood sample) obtained from a subject
within about
24 hours from the time point when the blood sample was drawn or obtained from
the subject
(but prior to testing or performance of an assay). In some embodiments, it is
advantageous to
process a blood sample obtained from a subject within about 12 hours, within
about 10 hours,
within about 8 hours, within about 6 hours, within about 4 hours, within about
2 hours, or
within about 1 hour from the time point when the blood sample was drawn from
the subject.
In other embodiments, it is advantageous to process a blood sample within a
time period after
the sample is obtained from a subject, including from about 1 hour to about 12
hours, from
about 1 hour to about 10 hours, from about 1 hour to about 8 hours, from about
1 hour to
about 6 hours, from about 1 hour to about 4 hours, or from about 1 hour to
about 2 hours,
after the sample has been obtained from the subject. In some embodiments, it
is
advantageous to process a blood sample obtained from a subject within about 12
hours or
less from the time point when the blood sample was drawn from the subject. In
some
embodiments, it is advantageous to process a blood sample obtained from a
subject within
about 10 hours or less from the time point when the blood sample was drawn
from the
subject. In some embodiments, it is advantageous to process a blood sample
obtained from a
subject within about 8 hours or less from the time point when the blood sample
was drawn
from the subject. In some embodiments, it is advantageous to process a blood
sample
obtained from a subject within about 6 hours or less from the time point when
the blood
sample was drawn from the subject. In some embodiments, it is advantageous to
process a
blood sample obtained from a subject within about 4 hours or less from the
time point when
the blood sample was drawn from the subject. In some embodiments, it is
advantageous to
process a blood sample obtained from a subject within about 2 hours or less
from the time
44

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
point when the blood sample was drawn from the subject. In some embodiments,
it is
advantageous to process a blood sample obtained from a subject within about 1
hour or less
from the time point when the blood sample was drawn from the subject.
101331 In accordance with these embodiments, it has been found that during
processing that
mechanical agitation (such as mixing) of the blood sample should be avoided
during or
between the period of time after the sample is obtained from the subject to
the time when the
test or assayed is performed (e.g., to detect and/or measure UCH-L1). In other
words,
continuous mixing up until testing or performance of an assay should be
avoided. In some
cases, mixing the blood samples during processing can cause hemolysis of the
blood samples
and release UCH-L1 into the sample, which can cause increased UCH-L1 levels to
be
obtained. However, one skilled in the art that would recognize that minor
mechanical
agitation or mixing may be necessary when processing blood samples; however,
mixing to a
degree that would lead to hemolysis should generally be avoided. For example,
a blood
sample should not be mixed for longer than is necessary to mix the blood
sample with any
anti-coagulant that may be present in a collection tube (e.g, such as in a
heparin tube,
ethylene diamine tetra acetic acid (EDTA) tube, etc.). In another aspect, the
blood sample
should not be mixed for longer than 5 seconds. In yet another aspect, the
blood sample
should not be mixed for longer than 10 seconds. In yet another aspect, the
blood sample
should not be mixed for longer than 15 seconds. In yet another aspect, the
blood sample
should not be mixed for longer than 20 seconds. In yet another aspect, the
blood sample
should not be mixed for longer than 25 seconds. In yet another aspect, the
blood sample
should not be mixed for longer than 30 seconds. In yet another aspect, the
blood sample
should not be mixed for longer than 40 seconds. In yet another aspect, the
blood sample
should not be mixed for longer than 50 seconds. In yet another aspect, the
blood sample
should not be mixed for longer than 60 seconds. In yet another aspect, the
blood sample
should not be mixed for longer than 2 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 3 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 4 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 5 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 5 minutes. In yet another aspect, the
blood sample

CA 03068041 2019-12-19
WO 2019/010131
PCT/US2018/040612
should not be mixed for longer than 2 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 3 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 4 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 5 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 6 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 7 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 8 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 9 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 10 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 11 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 12 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 13 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 14 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 15 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 16 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 17 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 18 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 19 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 20 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 25 minutes. In yet another aspect, the
blood sample
should not be mixed for longer than 30 minutes.
[0134]
[0135] In
accordance with these embodiments, in another aspect, the blood samples can be
maintained or adjusted at various temperatures during processing, including,
for example, at
temperatures ranging from about 2 C to about 8 C to about room temperature
(e.g., about
20 C to about 25 C). Blood samples can be maintained within these temperature
ranges for
any period of time prior to perfol ming a test or assay to assess (e.g.,
detect and/or measure)
UCH-L1 levels, including for up to about 24 hours after the sample has been
obtained from a
subject. In some cases, it is advantageous to maintain blood samples at a
temperature
ranging from about 2 C to about 8 C for no more than about 8 hours after the
sample has
46

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
been obtained from a subject, and up until the sample is assessed for UCH-L1
concentrations.
In some cases, it is advantageous to maintain blood samples at about room
temperature
(namely, from about 20 C to about 25 C) for no more than about 8 hours after
the sample
has been obtained from a subject, and up until the sample is assessed for UCH-
L1
concentrations. In some embodiments, whole blood assays (e.g., i-STATS and i-
STATS
Alinity) require working with samples in a manner that may include mixing or
prolonged
exposure to room temperature, which may have an effect on the assessment of
UCH-L1
concentrations, as described herein. In some embodiments, the blood samples
can be
maintained at a temperature from about 2 C to about 8 C. For example, if the
blood sample
is a whole sample, the sample can be maintained at a temperature of from about
2 C to about
8 C. By way of another example, if the blood sample is a plasma sample, the
sample can be
maintained at a temperature of from about 2 C to about 8 C. In yet another
example, if the
blood sample is a serum sample, the sample can be maintained at a temperature
of from
about 2 C to about 8 C. In another embodiment, the blood samples can be
maintained at
room temperature (e.g., about 20 C to about 25 C). For example, if the blood
sample is a
plasma sample, the sample can be maintained at room temperature. In yet
another example,
if the blood sample is a serum sample, the sample can be maintained at room
temperature.
[0136] Processing whole blood sample can include any steps or methods used
to obtain the
blood sample from the subject, as well as any steps or methods used to
separate out various
blood components into serum, plasma, and red blood cell (RBC) fractions.
Processing can
also include any steps or methods involved with testing, detecting, measuring,
or assessing
blood samples for various components, such as UCH-L1 and other biomarkers of
TBI. In
cases where assessing UCH-L1 levels are not included as part of processing the
whole blood
samples (e.g., UCH-L1 assessments are performed in a laboratory separate from
the site
where the blood samples were obtained from a subject), testing, detecting, or
measuring
UCH-L1 samples can occur shortly after or immediately after the whole blood
sample has
been processed. In some embodiments, methods of the present disclosure include
processing
a whole blood sample from a subject within no more than about 8 hours after
the sample is
obtained from the subject, in order to avoid a rise in UCH-L1 levels that
occurs due to
improper storage conditions and/or processing methods.
47

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0137] In some embodiments, processing includes separating serum and/or
plasma from blood
cells in the sample in order to assess UCH-Li levels in the plasma and/or
serum. This
process is generally referred to as blood fractionation and may be done by
subjecting the
blood samples to routine techniques known in the art, such as, for example,
centrifugation,
chromatography, ethanol fractionation, lab-on-a-chip cellular sorting (e.g.,
magnetic cellular
sorting), size exclusion in cross-flow filtration, dielectrophoretic
techniques, pinched-flow
fractionation, inertial microfluidics, separation utilizing electro-
hydrodynamic forces, and
combinations thereof.
[0138] In some embodiments, blood fractionation may be done by
centrifugation, which
results in the separation of the sample into a plasma component, a buffy coat
(leukocytes and
platelets), and erythrocytes (red blood cells). Collection tubes, which are
used to collect and
store blood samples obtained from subjects, can also be used to separate
plasma from blood
cells, such as during centrifugation. A variety of collection tubes can be
used in the methods
described herein. For example, collection tubes can contain or be coated to
contain various
components to facilitate the fractionation of blood components. In some
embodiments,
collection tubes can contain one or more anti-coagulants, such as EDTA) or
salts of EDTA
(such as K2EDTA or K3EDTA), which is an anti-coagulant that can be used to
prevent blood
clotting by chelating calcium, an essential component of coagulation.
Moreover, plasma
isolated from blood samples taken with EDTA collection tubes can also be used
to measure
or detect a variety of proteins found in the plasma, such as UCH-L1 and other
biomarkers of
TBI, and genetic material can easily be stored from buffy coats from EDTA
collection tubes.
Examples of other anti-coagulants that can be used include without limitation
heparin, such
as lithium or sodium heparin, ethylene glycol tetra acetic acid (EGTA), salts
of EGTA,
hirudin, heparin, citric acid, salts of citric acid, oxalic acid, salts of
oxalic acid (e.g.,
potassium oxalate, sodium oxalate, or the like), and combinations thereof.
[0139] In some embodiments, processing includes separating serum from
plasma and blood
cells in the sample in order to assess UCH-L1 levels in the serum. Serum
generally refers to
the blood component that remains after clotting occurs; serum is the cell-free
liquid that is
also depleted of coagulation factors. Serum can be separated from plasma and
blood cells in
the sample using the methods described previously herein. Specifically, serum
separation
48

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
tubes (SSTs) can be used to separate serum from blood cells (such as during
centrifugation).
Specifically, SSTs are tubes that may contain a gel, such as a silicone gel.
When centrifuged,
the silicone gel forms a layer on top of the buffy coat, allowing the blood
serum to be
removed more effectively for testing and related purposes. SSTs are sometimes
called
"marble-top tubes," "target-tops," or "gold-topped tubes," referring to the
stoppers which are
gold, red with a gold ring on top, or marbled red and grey. Marble-top tubes
are also referred
to as "tiger-tops" in some clinical settings.
[0140] The present disclosure is directed to an improvement of a method of
measuring an
amount of UCH-L1 in a blood sample, in which processing the sample includes
performing a
test that measures the amount of UCH-L1 in the sample. In some embodiments,
the test for
detecting, measuring, quantifying, or assessing UCH-L1 includes any available
methods for
assaying proteins, including, but not limited to, an immunoassay, chemical
analysis, SDS
PAGE and Western blot analysis, electrophoresis analysis, a protein assay, a
competitive
binding assay, a functional protein assay, chromatography, spectrophotometry,
and the like.
In some embodiments, immunoassays, such as the types described herein (e.g., i-
STAT and
i-STAT Alinity) are advantageous for measuring UCH-L1 levels in whole blood
samples.
[0141] In some embodiments, the present disclosure is directed to an
improvement of a
method of measuring an amount of UCH-L1 in a blood sample, which includes
performing
an immunoassay-based test for measuring UCH-Li. The immunoassay test can
include
contacting the sample, either simultaneously or sequentially, in any order
with at least one
capture antibody, which binds to an epitope on UCH-L1 or UCH-L1 fragment to
form a
capture antibody-UCH-L1 antigen complex. Additionally, the immunoassay can
include at
least one detection antibody, which includes a detectable label and binds to
an epitope on
UCH-L1 that is not bound by the capture antibody, to form a UCH-L1 antigen-
detection
antibody complex, such that a capture antibody-UCH-L1 antigen-detection
antibody complex
is formed. In some embodiments, measuring the amount or concentration of UCH-
L1 in a
sample is based on the signal generated by the detectable label in the capture
antibody-UCH-
Li antigen-detection antibody complex.
49

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
3. Methods of Aiding in the Diagnosis and Evaluation of Whether a Human
Subject has
Sustained an Injury to the Head
[0142] In another embodiment, the methods described herein can be used to
provide an aid in
the diagnosis and evaluation of whether a human subject has sustained or may
have sustained
an injury to the head by determining the levels of UCH-L1 in a subject. The
method can aid
in determining the extent of traumatic brain injury in a human subject with a
suspected injury
to the head, e.g., determining whether the subject has mild traumatic brain
injury or moderate
to severe traumatic brain injury. As used here, "determining whether the
subject has mild
traumatic brain injury or moderate to severe traumatic brain injury" refers to
the fact that the
aforementioned method can be used, e.g., with other information (e.g.,
clinical assessment
data), to determine that the subject is more likely than not to have mild
traumatic brain injury
or moderate to severe traumatic brain injury. The method may be used to detect
or assess
traumatic brain injury in a subject using the anti-UCH-L1 antibodies described
below, or
antibody fragments thereof. The method can include the steps of (a) obtaining
a sample from
a subject, (b) determining the level of UCH-L1 in the sample using one or more
anti-UCH-
Li antibodies, or antibody fragments thereof, and (c) determining whether the
subject has
sustained a mild or a moderate to severe traumatic brain injury (TBI) based on
the UCH-L1
levels. In some embodiments, the subject is determined as having (1) a
moderate, severe or
moderate to severe TBI when the level of the early biomarker in the sample is
higher than a
reference level of the early biomarker, or (2) a mild TBI when the level of
the early
biomarker in the sample is lower than a reference level of the early
biomarker. The sample
can be a blood sample.
[0143] Alternatively, the method can include the steps of (a) detei
mining the level of UCH-
Li in the sample obtained from a subject using one or more anti-UCH-L1
antibodies, or
antibody fragments thereof, and (b) determining whether the subject has
sustained a mild or a
moderate to severe traumatic brain injury (TBI) based on the UCH-L1 levels. In
some
embodiments, the subject is determined as having (1) a moderate, severe or
moderate to
severe TBI when the level of the early biomarker in the sample is higher than
a reference
level of the early biomarker, or (2) a mild FBI when the level of the early
biomarker in the

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
sample is lower than a reference level of the early biomarker. The sample can
be a blood
sample.
[0144] By measuring and assessing UCH-L1, the method allows for more diseases
to be more
accurately diagnosed and subsequently treated more successfully, compared to
other
commercially available UCH-L1 immunoassays. The method can be adapted for use
in an
automated system or a semi-automated system.
[0145] Generally, a reference level of UCH-L1 can be employed as a
benchmark against
which to assess results obtained upon assaying a test sample for UCH-Li.
Generally, in
making such a comparison, the reference level of UCH-L1 is obtained by running
a particular
assay a sufficient number of times and under appropriate conditions such that
a linkage or
association of analyte presence, amount or concentration with a particular
stage or endpoint
of TBI or with particular indicia can be made. Typically, the reference level
of UCH-L1 is
obtained with assays of reference subjects (or populations of subjects). The
UCH-L1
measured can include UCH-L1 fragments thereof, degradation products thereof,
and/or
enzymatic cleavage products thereof. In certain embodiments, the reference
level of UCH-
Li may be correlated with control subjects that have not sustained a head
injury. Generally,
the same assay used to determine the reference level is used to assay or test
the sample.
[0146] The reference level in this method can be the level of UCH-Li in a
subject having or
suspected of having traumatic brain injury. In some embodiments, levels higher
than or
equal to 5 pg/mL, 10 pg/mL, 20 pg/mL, 30 pg/mL, 40 pg/mL, 50 pg/mL, 60 pg/mL,
70
pg/mL, 80 pg/mL, 90 pg/mL, 100 pg/mL, 500 pg/mL, 1000 pg/mL, 5000 pg/mL, 10000
pg/mL, or 50000 pg/mL in serum of UCH-L1 identify the subject as having
traumatic brain
injury. Optionally, in some cases, levels higher than or equal to 100000
pg/mL, 500000
pg/mL, 1000000 pg/mL, 150000 pg/mL, 200000 pg/mL, or 500000 pg/mL in serum of
UCH-
Li identify the subject as having traumatic brain injury. It is known that,
absent
standardization, reference levels can vary between methods and assays.
Accordingly, the
method described herein is intended to apply to any appropriate reference
level.
[0147] In some embodiments, the method further includes treating the human
subject
predicted as having moderate to severe traumatic brain injury with a traumatic
brain injury
treatment, as described below. In some embodiments, the method further
includes
51

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
monitoring the human subject predicted as having mild traumatic brain injury,
as described
below.
4. Methods for Aiding in Determining the Risk of a Subject of Developing
Traumatic
Brain Injury
[0148] In yet another embodiment, the methods described herein also can be
used as an aid to
determine whether or not a subject has a traumatic brain injury or is at risk
of developing a
traumatic brain injury (such as a severe traumatic brain injury from a
moderate traumatic
brain injury) by determining the levels of UCH-Li in a subject using the anti-
UCH-L1
antibodies described below, or antibody fragments thereof. Thus, in particular
embodiments,
the disclosure also provides a method for aiding in determining whether a
subject having, or
at risk for, traumatic brain injuries, discussed herein and known in the art,
is a candidate for
therapy or treatment. As used here, "determining whether the subject has, or
is at risk for,
traumatic brain injuries refers to the fact that the aforementioned method can
be used, e.g.,
with other information (e.g., clinical assessment data), to determine that the
subject is more
likely than not to have traumatic brain injuries. Generally, the subject is at
least one who: (i)
has experienced, or is suspected of having experienced, an injury to the head;
(ii) ingested
and/or been exposed to one or more chemicals and/or toxins; (iii) suffers from
an
autoimmune disease, a metabolic disorder, a brain tumor, hypoxia, one or more
viruses,
meningitis, hydrocephalus or suffers from any combinations thereof; or (iv)
any
combinations of (i)-(iii); or, (v) who has actually been diagnosed as having,
or being at risk
for TBI (such as, for example, subjects suffering from an autoimmune disease,
a metabolic
disorder, a brain tumor, hypoxia, one or more viruses, meningitis,
hydrocephalus or
combinations thereof), and/or who demonstrates an unfavorable (i.e.,
clinically undesirable)
concentration or amount of UCH-L I or UCH-L1 fragment, as described herein.
[0149] Specifically, such a method can comprise the steps of: (a)
determining the
concentration or amount in a test sample from a subject of UCH-Li using the
methods
described herein, or methods known in the art); and (b) comparing the
concentration or
amount of UCH-Li determined in step (a) with a reference level, wherein, if
the
concentration or amount of UCH-L1 determined in step (a) is favorable with
respect to a
52

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
reference level, then the subject is determined not to have or be at risk for
traumatic brain
injury as discussed herein and known in the art. However, if the concentration
or amount of
UCH-L1 determined in step (a) is unfavorable with respect to the reference
level, then the
subject is determined to have or be at risk for traumatic brain injury as
discussed herein and
known in the art.
[0150] The reference level in this method can be the level of UCH-L1 in a
subject having or
suspected of having traumatic brain injury. In some embodiments, levels higher
than or
equal to 5 pg/mL, 10 pg/mL, 20 pg/mL, 30 pg/mL, 40 pg/mL, 50 pg/mL, 60 pg/mL,
70
pg/mL, 80 pg/mL, 90 pg/mL, 100 pg/mL, 500 pg/mL, 1000 pg/mL, 5000 pg/mL, 10000
pg/mL, or 50000 pg/mL in serum of UCH-L1 identify the subject as having
traumatic brain
injury. Optionally, in some cases, levels higher than or equal to 100000
pg/mL, 500000
pg/mL, 1000000 pg/mL, 150000 pg/mL, 200000 pg/mL, or 500000 pg/mL in serum of
UCH-
Li identify the subject as having traumatic brain injury. It is known that,
absent
standardization, reference levels can vary between methods and assays.
Accordingly, the
method described herein is intended to apply to any appropriate reference
level.
[0151] In some embodiments, the method further includes treating the human
subject with a
traumatic brain injury with a traumatic brain injury treatment and/or
monitoring the human
subject, as described below.
5. Method of Aiding in the Determination of Whether to Perform a CT scan on a
Human
Subject Who has Sustained an Injury to the Head
[0152] The present disclosure relates, among other methods, to a method of
aiding in
determining whether to perform a computerized tomography (CT) scan on a human
subject
who has sustained or may have sustained a suspected injury to the head. As
used here,
"determination of whether to perform a CT scan on a human subject" refers to
the fact that
the aforementioned method can be used, e.g., with other infoimation (e.g.,
clinical
assessment data), to determine that the subject is more likely than not to
have a positive head
CT scan. Specifically, such a method can comprise the steps of: (a) performing
an assay on a
sample obtained from the subject within about 24 hours after a suspected
injury to the head to
measure or detect a level of UCH-L1 in the sample; and (b) performing a CT
scan on the
53

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
subject when the level of UCH-L1 in the sample is higher than a reference
level of UCH-L1
and not perfoiming a CT scan on the subject when the level of UCH-L1 in the
sample is
lower than a reference level of UCH-Ll. The sample can be a sample.
[0153] In this aspect of the invention, one skilled in the art would
appreciate the blood
samples obtained from such subjects would be processed using the methods as
described
previously herein as quickly as possible (such as, for example, in about 8
hours or less) to
allow a test or assay to be performed within about 24 hours after a suspected
injury to the
head as described herein.
[0154] By measuring and assessing UCH-L1, the method allows for the need to
perform a CT
scan on a subject more accurately, and subsequently treated more successfully,
compared to
other commercially available UCH-L1 immunoassays. The method can be adapted
for use in
an automated system or a semi-automated system.
[0155] Generally, a reference level of UCH-L1 can be employed as a
benchmark against
which to assess results obtained upon assaying a test sample for UCH-Li.
Generally, in
making such a comparison, the reference level of UCH-L1 is obtained by running
a particular
assay a sufficient number of times and under appropriate conditions such that
a linkage or
association of analyte presence, amount or concentration with a particular
stage or endpoint
of TBI or with particular indicia can be made. Typically, the reference level
of UCH-L1 is
obtained with assays of reference subjects (or populations of subjects). The
UCH-L1
measured can include UCH-L1 fragments thereof, degradation products thereof,
and/or
enzymatic cleavage products thereof. In certain embodiments, the reference
level of UCH-
Li may be correlated with control subjects that have not sustained a head
injury.
[0156] The reference level in this method can be the level of UCH-L1 in a
subject having or
suspected of having traumatic brain injury. In some embodiments, levels higher
than or
equal to 5 pg/mL, 10 pg/mL, 20 pg/mL, 30 pg/mL, 40 pg/mL, 50 pg/mL, 60 pg/mL,
70
pg/mL, 80 pg/mL, 90 pg/mL, 100 pg/mL, 500 pg/mL, 1000 pg/mL, 5000 pg/mL, 10000
pg/mL, or 50000 pg/mL in serum of UCH-L1 identify the subject as having
traumatic brain
injury. Optionally, in some cases, levels higher than or equal to 100000
pg/mL, 500000
pg/mL, 1000000 pg/mL, 150000 pg/mL, 200000 pg/mL, or 500000 pg/mL in serum of
UCH-
Li identify the subject as having traumatic brain injury. It is known that,
absent
54

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
standardization, reference levels can vary between methods and assays.
Accordingly, the
method described herein is intended to apply to any appropriate reference
level.
[0157] In some embodiments, the method further includes treating the human
subject with a
traumatic brain injury with a traumatic brain injury treatment and/or
monitoring the human
subject, as described below.
6. Methods of Aiding in Determining the Extent of Traumatic Brain Injury in a
Human
Subject Who May have Sustained an Injury to the Head
[0158] The present disclosure relates to a method of aiding in determining
the extent of
traumatic brain injury in a human subject with a suspected injury to the head,
e.g.,
determining whether the subject has mild traumatic brain injury or moderate to
severe
traumatic brain injury. As used here, "detellnining the extent of traumatic
brain injury in a
human subject with a suspected injury to the head" refers to the fact that the
aforementioned
method can be used, e.g., with other information (e.g., clinical assessment
data), to determine
that the subject is more likely than not to have mild traumatic brain injury
or moderate to
severe traumatic brain injury. The method includes performing an assay on at
least two
samples obtained from the subject and detecting UCH-L1 in the at least two
samples. The
first sample is taken from the human subject within 24 hours of injury and the
second sample
is taken from the human subject about 3 to about 6 hours after the first
sample is taken. The
UCH-L1 appears within about 2 to about 24 hours after the onset of injury to
the head. The
onset of the presence of UCH-LI appears within about 0 to about 6 hours after
the onset of
the suspected injury. Levels of UCH-L1 are determined for each of the first
sample and
second sample. The level of UCH-Li is determined to decrease or increase. The
extent of
the traumatic brain injury is determined in the subject based on whether the
level of UCH-L1
decreases, increases, or remains the same from the first sample to the second
sample. The
UCH-LI increases within about 0 to about 6 hours after the suspected injury
and then
decreases or increases thereafter in subjects with traumatic brain injury. In
some
embodiments, the onset of the presence of UCH-Li appears within about 0, about
0.5 hours,
about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, or
about 6 hours
after injury to the head.

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0159] In this aspect of the invention, one skilled in the art would
appreciate the blood
samples obtained from such subjects would be processed using the methods as
described
previously herein as quickly as possible (such as, for example, in about 8
hours or less) to
allow a test or assay to be performed within about 24 hours after a suspected
injury to the
head as described herein.
[0160] In some embodiments, the first sample is taken from the subject at a
first time point
within 24 hours of the suspected injury and the second sample is taken from
the subject at a
second time point after the first time point and the subject is determined to
have mild or mild
to severe traumatic brain injury when the level of UCH-L1 decreases from the
first sample to
the second sample. In some embodiments, the UCH-L1 decreases at least about 5%
from the
increased levels. For example, the UCH-L1 levels may decrease about 5%, about
10%, about
20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about
90%,
about 100%, about 200%, about 300%, about 400%, about 500%, about 600%, about
700%,
about 800%, about 900%, or about 1000% from the increased levels. In some
embodiments,
the UCH-L1 decreases at least about 0.1-fold, at least about 0.2-fold, at
least about 0.3-fold,
at least about 0.4-fold, at least about 0.5-fold, at least about 0.55-fold, at
least about 0.6-fold,
at least about 0.7-fold, at least about 0.73-fold, at least about 0.8-fold, at
least about 0.9-fold,
at least about 1-fold, at least about 1.5-fold, at least about 2-fold, at
least about 3-fold, at least
about 4-fold, at least about 5-fold, at least about 6-fold, at least about 7-
fold, at least 8-fold,
at least 9-fold, at least 10-fold, at least 11-fold, at least 12-fold, at
least 13-fold, at least 14-
fold, at least 15-fold, at least 16-fold, at least 17-fold, at least 18-fold,
at least 19-fold, or at
least 20-fold from the increased levels. In some embodiments, the UCH-L1
decreases less
than about 0.1-fold, less than about 0.2-fold, less than about 0.3-fold, less
than about 0.4-
fold, less than about 0.5-fold, less than about 0.55-fold, at least about 0.6-
fold, at least about
0.7-fold, at least about 0.73-fold, at least about 0.8-fold, at least about
0.9-fold, less than
about 1-fold, less than about 1.5-fold, less than about 2-fold, less than
about 3-fold, less than
about 4-fold, less than about 5-fold, less than about 6-fold, less than about
7-fold, less than 8-
fold, less than 9-fold, less than 10-fold, less than 11-fold, less than 12-
fold, less than 13-fold,
less than 14-fold, less than 15-fold, less than 16-fold, less than 17-fold,
less than 18-fold, less
than 19-fold, or less than 20-fold from the increased levels.
56

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0161] In some embodiments, the second sample is obtained within about 1
hour to about 10
hours after the first sample is obtained, such as about 3 hours to about 6
hours after the first
sample is obtained. In some embodiments, the second time sample is obtained
within about
1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6
hours, about 7
hours, about 8 hours, about 9 hours, or about 10 hours after the first sample
is obtained.
[0162] In some embodiments, the method further includes treating a human
subject assessed
as having moderate to severe traumatic brain injury with a traumatic brain
injury treatment,
as described below. In some embodiments, the method further includes
monitoring a human
subject assessed as having mild traumatic brain injury, as described below.
7. Methods of Monitoring the Progression of Traumatic Brain Injury in a
Subject
[0163] In yet another embodiment, the methods described herein also can be
used as an aid to
monitor the progression of disease and/or injury, such as traumatic brain
injury, in a subject
by determining the levels of UCH-L1 in a subject using the anti-UCH-L1
antibodies
described below, or antibody fragments thereof As used here, "monitoring the
progression
of disease and/or injury" refers to the fact that the aforementioned method
can be used, e.g.,
with other information (e.g., clinical assessment data), to determine if the
disease in the
subject has more likely than not continued, progressed or worsened. Optimally,
the method
includes the steps of (a) determining the concentration or amount of UCH-L1 in
a test sample
from a subject using the anti-UCH-L1 antibodies described below, or antibody
fragments
thereof, (b) determining the concentration or amount of UCH-L1 in a later test
sample from a
subject using the anti-UCH-L1 antibodies described below, or antibody
fragments thereof,
and (c) comparing the concentration or amount of UCH-L1 as determined in step
(b) with the
concentration or amount of UCH-L1 determined in step (a), wherein if the
concentration or
amount determined in step (b) is unchanged or is unfavorable when compared to
the
concentration or amount of UCH-L1 detemiined in step (a), then the disease in
the subject is
determined to have continued, progressed or worsened. By comparison, if the
concentration
or amount of UCH-L1 as determined in step (b) is favorable when compared to
the
concentration or amount of UCH-L1 as determined in step (a), then the disease
in the subject
is determined to have discontinued, regressed or improved.
57

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0164] Optionally, the method further comprises comparing the concentration
or amount of
UCH-L1 as determined in step (b), for example, with a reference level.
Further, optionally
the method comprises treating the subject with one or more pharmaceutical
compositions for
a period of time if the comparison shows that the concentration or amount of
UCH-L1 as
determined in step (b), for example, is unfavorably altered with respect to
the reference level.
[0165] Still further, the methods can be used to monitor treatment in a
subject receiving
treatment with one or more pharmaceutical compositions or other traumatic
brain injury
treatment, as described below. Specifically, such methods involve providing a
first test
sample from a subject before the subject has been administered one or more
pharmaceutical
compositions. Next, the concentration or amount in a first test sample from a
subject of
UCH-L1 is determined (e.g., using the methods described herein or as known in
the art).
After the concentration or amount of UCH-L1 is determined, optionally the
concentration or
amount of UCH-L1 is then compared with a reference level. If the concentration
or amount
of UCH-L1 as determined in the first test sample is lower than the reference
level, then the
subject is not treated with one or more pharmaceutical compositions or
alternatively, the
subject may be treated with one or more pharmaceutical compositions. If the
concentration or
amount of UCH-L1 as determined in the first test sample is higher than the
reference level,
then the subject is treated with one or more phaimaceutical compositions for a
period of time
or alternatively, the subject is not treated with one or more pharmaceutical
compositions. The
period of time that the subject is treated with the one or more pharmaceutical
compositions
can be determined by one skilled in the art (for example, the period of time
can be from
about seven (7) days to about two years, preferably from about fourteen (14)
days to about
one (1) year).
[0166] During the course of treatment with the one or more pharmaceutical
compositions,
second and subsequent test samples are then obtained from the subject. The
number of test
samples and the time in which said test samples are obtained from the subject
are not critical.
For example, a second test sample could be obtained seven (7) days after the
subject is first
administered the one or more pharmaceutical compositions, a third test sample
could be
obtained two (2) weeks after the subject is first administered the one or more
pharmaceutical
compositions, a fourth test sample could be obtained three (3) weeks after the
subject is first
58

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
administered the one or more pharmaceutical compositions, a fifth test sample
could be
obtained four (4) weeks after the subject is first administered the one or
more pharmaceutical
compositions, etc.
[0167] After each second or subsequent test sample is obtained from the
subject, the
concentration or amount of UCH-L1 is determined in the second or subsequent
test sample is
determined (e.g., using the methods described herein or as known in the art).
The
concentration or amount of UCH-L1 as determined in each of the second and
subsequent test
samples is then compared with the concentration or amount of UCH-L1 as
determined in the
first test sample (e.g., the test sample that was originally optionally
compared to the reference
level). If the concentration or amount of UCH-L1 as determined in step (c) is
favorable when
compared to the concentration or amount of UCH-L1 as determined in step (a),
then the
disease in the subject is determined to have discontinued, regressed, or
improved, and the
subject can continue to be administered the one or pharmaceutical compositions
of step (b).
However, if the concentration or amount determined in step (c) is unchanged or
is
unfavorable when compared to the concentration or amount of UCH-L1 as
determined in
step (a), then the disease in the subject is determined to have continued,
progressed or
worsened, and the subject can be treated with a higher concentration of the
one or more
pharmaceutical compositions administered to the subject in step (b) or the
subject can be
treated with one or more pharmaceutical compositions that are different from
the one or more
pharmaceutical compositions administered to the subject in step (b).
Specifically, the subject
can be treated with one or more pharmaceutical compositions that are different
from the one
or more pharmaceutical compositions that the subject had previously received
to decrease or
lower said subject's UCH-L1 level.
[0168] Generally, for assays in which repeat testing may be done (e.g.,
monitoring disease
progression and/or response to treatment), a second or subsequent test sample
is obtained at a
period in time after the first test sample has been obtained from the subject.
Specifically, a
second test sample from the subject can be obtained minutes, hours, days,
weeks or years
after the first test sample has been obtained from the subject. For example,
the second test
sample can be obtained from the subject at a time period of about 1 minute,
about 5 minutes,
about 10 minutes, about 15 minutes, about 30 minutes, about 45 minutes, about
60 minutes,
59

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours,
about 7 hours,
about 8 hours, about 9 hours, about 10 hours, about 11 hours, about 12 hours,
about 13 hours,
about 14 hours, about 15 hours, about 16 hours, about 17 hours, about 18
hours, about 19
hours, about 20 hours, about 21 hours, about 22 hours, about 23 hours, about
24 hours, about
2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days,
about 2 weeks,
about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks,
about 8 weeks,
about 9 weeks, about 10 weeks, about 11 weeks, about 12 weeks, about 13 weeks,
about 14
weeks, about 15 weeks, about 16 weeks, about 17 weeks, about 18 weeks, about
19 weeks,
about 20 weeks, about 21 weeks, about 22 weeks, about 23 weeks, about 24
weeks, about 25
weeks, about 26 weeks, about 27 weeks, about 28 weeks, about 29 weeks, about
30 weeks,
about 31 weeks, about 32 weeks, about 33 weeks, about 34 weeks, about 35
weeks, about 36
weeks, about 37 weeks, about 38 weeks, about 39 weeks, about 40 weeks, about
41 weeks,
about 42 weeks, about 43 weeks, about 44 weeks, about 45 weeks, about 46
weeks, about 47
weeks, about 48 weeks, about 49 weeks, about 50 weeks, about 51 weeks, about
52 weeks,
about 1.5 years, about 2 years, about 2.5 years, about 3.0 years, about 3.5
years, about 4.0
years, about 4.5 years, about 5.0 years, about 5.5. years, about 6.0 years,
about 6.5 years,
about 7.0 years, about 7.5 years, about 8.0 years, about 8.5 years, about 9.0
years, about 9.5
years, or about 10.0 years after the first test sample from the subject is
obtained.
[0169] When used to monitor disease progression, the above assay can be
used to monitor the
progression of disease in subjects suffering from acute conditions. Acute
conditions, also
known as critical care conditions, refer to acute, life-threatening diseases
or other critical
medical conditions involving, for example, the cardiovascular system or
excretory system.
Typically, critical care conditions refer to those conditions requiring acute
medical
intervention in a hospital-based setting (including, but not limited to, the
emergency room,
intensive care unit, trauma center, or other emergent care setting) or
administration by a
paramedic or other field-based medical personnel. For critical care
conditions, repeat
monitoring is generally done within a shorter time frame, namely, minutes,
hours or days
(e.g., about 1 minute, about 5 minutes, about 10 minutes, about 15 minutes,
about 30
minutes, about 45 minutes, about 60 minutes, about 2 hours, about 3 hours,
about 4 hours,
about 5 hours, about 6 hours, about 7 hours, about 8 hours, about 9 hours,
about 10 hours,

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
about 11 hours, about 12 hours, about 13 hours, about 14 hours, about 15
hours, about 16
hours, about 17 hours, about 18 hours, about 19 hours, about 20 hours, about
21 hours, about
22 hours, about 23 hours, about 24 hours, about 2 days, about 3 days, about 4
days, about 5
days, about 6 days or about 7 days), and the initial assay likewise is
generally done within a
shorter timeframe, e.g., about minutes, hours or days of the onset of the
disease or condition.
10170] The assays also can be used to monitor the progression of disease in
subjects suffering
from chronic or non-acute conditions. Non-critical care conditions or non-
acute conditions,
refers to conditions other than acute, life-threatening disease or other
critical medical
conditions involving, for example, the cardiovascular system and/or excretory
system.
Typically, non-acute conditions include those of longer-term or chronic
duration. For non-
acute conditions, repeat monitoring generally is done with a longer timeframe,
such as hours,
days, weeks, months or years (e.g., about 1 hour, about 2 hours, about 3
hours, about 4 hours,
about 5 hours, about 6 hours, about 7 hours, about 8 hours, about 9 hours,
about 10 hours,
about 11 hours, about 12 hours, about 13 hours, about 14 hours, about 15
hours, about 16
hours, about 17 hours, about 18 hours, about 19 hours, about 20 hours, about
21 hours, about
22 hours, about 23 hours, about 24 hours, about 2 days, about 3 days, about 4
days, about 5
days, about 6 days, about 7 days, about 2 weeks, about 3 weeks, about 4 weeks,
about 5
weeks, about 6 weeks, about 7 weeks, about 8 weeks, about 9 weeks, about 10
weeks, about
11 weeks, about 12 weeks, about 13 weeks, about 14 weeks, about 15 weeks,
about 16
weeks, about 17 weeks, about 18 weeks, about 19 weeks, about 20 weeks, about
21 weeks,
about 22 weeks, about 23 weeks, about 24 weeks, about 25 weeks, about 26
weeks, about 27
weeks, about 28 weeks, about 29 weeks, about 30 weeks, about 31 weeks, about
32 weeks,
about 33 weeks, about 34 weeks, about 35 weeks, about 36 weeks, about 37
weeks, about 38
weeks, about 39 weeks, about 40 weeks, about 41 weeks, about 42 weeks, about
43 weeks,
about 44 weeks, about 45 weeks, about 46 weeks, about 47 weeks, about 48
weeks, about 49
weeks, about 50 weeks, about 51 weeks, about 52 weeks, about 1.5 years, about
2 years,
about 2.5 years, about 3.0 years, about 3.5 years, about 4.0 years, about 4.5
years, about 5.0
years, about 5.5, years, about 6.0 years, about 6.5 years, about 7.0 years,
about 7.5 years,
about 8.0 years, about 8.5 years, about 9.0 years, about 9.5 years or about
10.0 years), and the
61

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
initial assay likewise generally is done within a longer time frame, e.g.,
about hours, days,
months or years of the onset of the disease or condition.
[0171] Furthermore, the above assays can be performed using a first test
sample obtained
from a subject where the first test sample is obtained from one source, such
as whole blood,
serum, or plasma. Optionally the above assays can then be repeated using a
second test
sample obtained from the subject where the second test sample is obtained from
another
source. For example, if the first test sample was obtained from whole blood,
the second test
sample can be obtained from serum or plasma. The results obtained from the
assays using the
first test sample and the second test sample can be compared. The comparison
can be used to
assess the status of a disease or condition in the subject.
[0172] With respect to a reference level as employed for monitoring disease
progression
and/or treatment or for determining the risk of a subject of developing
traumatic brain injury,
the amount or concentration of UCH-L1 or UCH-L1 fragment may be "unchanged,"
"favorable" (or "favorably altered"), or "unfavorable" (or "unfavorably
altered"). "Elevated"
or "increased" refers to an amount or a concentration in a test sample that is
higher or greater
than a typical or normal level or range (e.g., reference level), or is higher
or greater than
another reference level or range (e.g., earlier or baseline sample). The term
"lowered" or
"reduced" refers to an amount or a concentration in a test sample that is
lower or less than a
typical or normal level or range (e.g., reference level), or is lower or less
than another
reference level or range (e.g., earlier or baseline sample). The term
"altered" refers to an
amount or a concentration in a sample that is altered (increased or decreased)
over a typical
or normal level or range (e.g., reference level), or over another reference
level or range (e.g.,
earlier or baseline sample).
[0173] The typical or normal level or range for UCH-L1 is defined in
accordance with
standard practice. A so-called altered level or alteration can be considered
to have occurred
when there is any net change as compared to the typical or normal level or
range, or
reference level or range that cannot be explained by experimental error or
sample variation.
Thus, the level measured in a particular sample will be compared with the
level or range of
levels determined in similar samples from a so-called normal subject. In this
context, a
"normal subject" is an individual with no detectable disease or disorder, and
a "normal"
62

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
(sometimes termed "control") patient or population is/are one(s) that
exhibit(s) no detectable
disease or disorder, respectively, for example. An "apparently normal subject"
is one in
which UCH-L1 has not been or is being assessed. The level of an analyte is
said to be
"elevated" when the analyte is normally undetectable (e.g., the normal level
is zero, or within
a range of from about 25 to about 75 percentiles of normal populations), but
is detected in a
test sample, as well as when the analyte is present in the test sample at a
higher than normal
level. Thus, inter alia, the disclosure provides a method of screening for a
subject having, or
at risk of having, traumatic brain injury.
8. Methods of Diagnosing or Assessing Involving Other Factors
[0174] The methods of diagnosing, prognosticating, and/or assessing, as
described above, can
further include using other factors for the diagnosis, prognostication, and
assessment. In
some embodiments, traumatic brain injury may be diagnosed using the Glasgow
Coma Scale
or the outcome of the traumatic brain injury may be predicted using the
Extended Glasgow
Outcome Scale (GOSE). Other tests, scales or indices can also be used either
alone or in
combination with the Glasgow Coma Scale. An example is the Ranchos Los Amigos
Scale.
The Ranchos Los Amigos Scale measures the levels of awareness, cognition,
behavior and
interaction with the environment. The Ranchos Los Amigos Scale includes: Level
I: No
Response; Level II: Generalized Response; Level III: Localized Response; Level
IV:
Confused-agitated; Level V: Confused-inappropriate; Level VI: Confused-
appropriate; Level
VII: Automatic-appropriate; and Level VIII: Purposeful-appropriate.
[0175] Other classification systems based on CT scan results can be used to
predict outcome
in patients, such as any classification systems known in the art. An example
is the Marshall
classification of traumatic brain injury, which places patients into one of
six categories (Ito
VI) of increasing severity on the basis of findings on non-contrast CT scan of
the brain.
Higher categories have worse prognosis and survival. The Marshall
classification is
primarily concerned with two features: 1) degree of swelling, as determined by
midline shift
and/or compression of basal cisterns, and 2) presence and size of
contusions/hemorrhages
referred to "high or mixed density lesions." Another example is the Rotterdam
score, which
incorporates additional variables (e.g. subarachnoid hemorrhage) and attempts
to address
63

some of the recognized limitations of the Marshall system, such as struggling
to classifying
patients who have injuries of multiple types. The Rotterdam classification
includes four
independently scored elements. Similar to the Marshall system, the Rotterdam
classification
includes 1) degree of basal cistern compression and 2) degree of midline
shift. The
Rotterdam does not, however, include contusions, but rather restricts mass
lesions to 3)
epidural hematomas, and adds 4) intraventricular and/or subarachnoid blood.
Each of these
is given a score, and these scores are tallied, with the addition of 1 to the
total. Higher scores
worse prognosis and survival.
[0176] Some instruments (such as, for example the Abbott Laboratories
instrument
ARCHITECT , and other core laboratory instruments) other than a point-of-care
device may
be capable of measuring levels of UCH-L1 in a sample higher or greater than
25,000 pg/mL.
Use of the methods as described herein may provide one or more of the benefits
described
herein on those devices (e.g., measure up to 25,000 pg/mL, dynamic range of 5
log, assay
linearity over the dynamic range, measure of UCH-L1 in a volume less than 20
microliters of
sample, expanded window of detection, etc.).
[0177] Other methods of detection include the use of or can be adapted for use
on a nanopore
device or nanowell device. Examples of nanopore devices are described in
International
Patent Publication No. WO 2016/161402.
Examples of nanowell device are described in International Patent Publication
No.
WO 2016/161400.
9. Combinations of UCH-L1 with other Biomarkers
[0178] The antibodies described herein can be used in a variety of methods
to detect and
measure levels and concentrations of UCH-L1 in combination with one or more
biomarkers
or immunoassays specific for disease. The present disclosure contemplates that
the
combination of UCH-Li with one or more biomarkers or immunoassays specific for
disease
may provide a greater discrimination between healthy controls and individuals
with disease
compared to measuring UCH-1A alone. For example, measuring a panel of UCH-L1
and
additional traumatic brain injury biomarkers may provide a greater
discrimination between
healthy controls and individuals with disease compared to a panel of UCH-Li
alone. The
64
Date Recue/Date Received 2023-06-29

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
combination of UCH-L1 with at least one or more biomarkers may provide greater
discrimination between healthy controls and individuals who have traumatic
brain injury.
[0179] Examples of the one or more biomarkers include glial fibrillary
acidic protein (GFAP),
S100 calcium-binding protein B (S100b), brain lipid binding protein (BLBP),
aldolase C
(ALDOC), astrocytic phosphoprotein 15 (PEA15), glutamine synthetase (GS), and
crystallin
B chain (CRYAB).
10. Treatment and Monitoring of Subjects Suffering from Traumatic Brain Injury
[0180] The subject identified or assessed in the methods described above as
having traumatic
brain injury, such as mild traumatic brain injury or moderate to severe
traumatic brain injury,
may be treated or monitored. In some embodiments, the method further includes
treating the
human subject assessed as having traumatic brain injury with a traumatic brain
injury
treatment, such as any treatments known in the art. For example, treatment of
traumatic
brain injury can take a variety of forms depending on the severity of the
injury to the head.
For example, for subjects suffering from mild TBI, the treatment may include
one or more of
rest, abstaining from physical activities, such as sports, avoiding light or
wearing sunglasses
when out in the light, medication for relief of a headache or migraine, anti-
nausea
medication, etc. Treatment for patients suffering from severe TBI might
include
administration of one or more appropriate medications (such as, for example,
diuretics, anti-
convulsant medications, medications to sedate and put an individual in a drug-
induced coma,
or other pharmaceutical or biopharmaceutical medications (either known or
developed in the
future for treatment of TBI), one or more surgical procedures (such as, for
example, removal
of a hematoma, repairing a skull fracture, decompressive craniectomy, etc.)
and one or more
therapies (such as, for example one or more rehabilitation, cognitive
behavioral therapy,
anger management, counseling psychology, etc.). In some embodiments, the
method further
includes monitoring the human subject assessed as having traumatic brain
injury (e.g., mild
or moderate to severe traumatic). In some embodiments, a subject identified as
having
traumatic brain injury, such as mild traumatic brain injury or severe
traumatic brain injury,
may be monitored with CT scan or MRI.

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
11. Methods of Measuring UCH-L1
[0181] In the methods described above, UCH-Li levels can be measured by any
means, such
as antibody dependent methods, such as immunoassays, protein
immunoprecipitation,
immunoelectrophoresis, chemical analysis, SDS-PAGE and Western blot analysis,
protein
immunostaining, electrophoresis analysis, a protein assay, a competitive
binding assay, a
functional protein assay, or chromatography or spectrometry methods, such as
high-
performance liquid chromatography (HPLC) or liquid chromatography¨mass
spectrometry
(LC/MS). Also, the assay can be employed in clinical chemistry format such as
would be
known by one skilled in the art. For example, a clinical chemistry format can
include an
assay that involves one antibody or no antibody. Examples of analyzers that
can be used for
the clinical chemistry format are described in U.S. Patent publication Nos.
2016/0320422 and
2015/0112630.
[0182] In some embodiments, measuring the level of UCH-Li includes
contacting the sample
with a first specific binding member and second specific binding member. In
some
embodiments the first specific binding member is a capture antibody and the
second specific
binding member is a detection antibody. In some embodiments, measuring the
level of UCH-
Li includes contacting the sample, either simultaneously or sequentially, in
any order: (1) a
capture antibody (e.g., UCH-Li -capture antibody), which binds to an epitope
on UCH-L1 or
UCH-L1 fragment to form a capture antibody-UCH-L1 antigen complex (e.g., UCH-
Li-
capture antibody-UCH-L1 antigen complex), and (2) a detection antibody (e.g.,
UCH-Li-
detection antibody), which includes a detectable label and binds to an epitope
on UCH-L1
that is not bound by the capture antibody, to form a UCH-L1 antigen-detection
antibody
complex (e.g., UCH-L1 antigen-UCH-Li-detection antibody complex), such that a
capture
antibody-UCH-L1 antigen-detection antibody complex (e.g., UCH-L1-capture
antibody-
UCH-L1 antigen-UCH-Li-detection antibody complex) is formed, and measuring the
amount or concentration of UCH-L1 in the sample based on the signal generated
by the
detectable label in the capture antibody-UCH-L1 antigen-detection antibody
complex.
[0183] In some embodiments, the first specific binding member is
immobilized on a solid
support. In some embodiments, the second specific binding member is
immobilized on a
66

solid support. In some embodiments, the first specific binding member is a UCH-
Li
antibody as described below.
[0184] In some embodiments, the sample is diluted or undiluted. The sample can
be from
about 1 to about 25 microliters, about 1 to about 24 microliters, about 1 to
about 23
microliters, about 1 to about 22 microliters, about 1 to about 21 microliters,
about 1 to about
20 microliters, about 1 to about 18 microliters, about 1 to about 17
microliters, about 1 to
about 16 microliters, about 15 microliters or about 1 microliter, about 2
microliters, about 3
microliters, about 4 microliters, about 5 microliters, about 6 microliters,
about 7 microliters,
about 8 microliters, about 9 microliters, about 10 microliters, about 11
microliters, about 12
microliters, about 13 microliters, about 14 microliters, about 15 microliters,
about 16
microliters, about 17 microliters, about 18 microliters, about 19 microliters,
about 20
microliters, about 21 microliters, about 22 microliters, about 23 microliters,
about 24
microliters or about 25 microliters. In some embodiments, the sample is from
about 1 to
about 150 microliters or less or from about 1 to about 25 microliters or less.
[0185] Some instruments (such as, for example the Abbott Laboratories
instrument
ARCHITECT , and other core laboratory instruments) other than a point-of-care
device may
be capable of measuring levels of UCH-L1 in a sample higher or greater than
25,000 pg/mL.
[0186] Other methods of detection include the use of or can be adapted for use
on a nanopore
device or nanowell device. Examples of nanopore devices are described in
International
Patent Publication No. WO 2016/161402.
Examples of nanowell device are described in International Patent Publication
No.
WO 2016/161400.
12. Ubiquitin Carboxy-Terminal Hydrolase Li (UCH-L1) Antibodies
[0187] The methods described herein may use an isolated antibody that
specifically binds to
ubiquitin carboxy-terminal hydrolase Li ("UCH-L1") (or fragments thereof),
referred to as
"UCH-Li antibody." The UCH-Li antibodies can be used to assess the UCH-L1
status as a
measure of traumatic brain injury, detect the presence of UCH-L1 in a sample,
quantify the
amount of UCH-LI present in a sample, or detect the presence of and quantify
the amount of
UCH-L1 in a sample.
67
Date Recue/Date Received 2023-06-29

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
a) Ubiquitin Carboxy-Terminal Hydrolase Li (UCH-L1)
[0188] Ubiquitin carboxy-terminal hydrolase Li ("UCH-L1"), which is also
known as
"ubiquitin C-terminal hydrolase," is a deubiquitinating enzyme. UCH-Li is a
member of a
gene family whose products hydrolyze small C-terminal adducts of ubiquitin to
generate the
ubiquitin monomer. Expression of UCH-L1 is highly specific to neurons and to
cells of the
diffuse neuroendocrine system and their tumors. It is abundantly present in
all neurons
(accounts for 1-2% of total brain protein), expressed specifically in neurons
and testis/ovary.
The catalytic triad of UCH-L1 contains a cysteine at position 90, an aspartate
at position 176,
and a histidine at position 161 that are responsible for its hydrolase
activity.
[0189] Human UCH-L1 may have the following amino acid sequence:
MQLKPMEINPEMLNKVLSRLGVAGQWRFVDVLGLEEESLGSVPAPACALLLLFPLT
AQHENFRICKQIEELKGQEVSPKVYFMKQTIGNSCGTIGLIHAVANNQDKLGFEDGSV
LKQFLSETEKMSPEDRAKCFEKNEATQAAHDAVAQEGQCRVDDKVNFHFILFNNVD
GHLYELDGRMPFPVNHGASSEDTLLKDAAKVCREFTEREQGEVRF SAVALCKAA
(SEQ ID NO: 1). The human UCH-L1 may be a fragment or variant of SEQ ID NO: 1.
The
fragment of UCH-L1 may be between 5 and 225 amino acids, between 10 and 225
amino
acids, between 50 and 225 amino acids, between 60 and 225 amino acids, between
65 and
225 amino acids, between 100 and 225 amino acids, between 150 and 225 amino
acids,
between 100 and 175 amino acids, or between 175 and 225 amino acids in length.
The
fragment may comprise a contiguous number of amino acids from SEQ ID NO: 1.
b) UCH-L1-Recognizing Antibody
[0190] The antibody is an antibody that binds to UCH-L1, a fragment
thereof, an epitope of
UCH-L1, or a variant thereof The antibody may be a fragment of the anti-UCH-L1
antibody
or a variant or a derivative thereof. The antibody may be a polyclonal or
monoclonal
antibody. The antibody may be a chimeric antibody, a single chain antibody, an
affinity
matured antibody, a human antibody, a humanized antibody, a fully human
antibody or an
antibody fragment, such as a Fab fragment, or a mixture thereof. Antibody
fragments or
derivatives may comprise F(ab')2, Fv or scFv fragments. The antibody
derivatives can be
68

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
produced by peptidomimetics. Further, techniques described for the production
of single
chain antibodies can be adapted to produce single chain antibodies.
[0191] The anti-UCH-L1 antibodies may be a chimeric anti-UCH-Li or
humanized anti-
UCH-Li antibody. In one embodiment, both the humanized antibody and chimeric
antibody
are monovalent. In one embodiment, both the humanized antibody and chimeric
antibody
comprise a single Fab region linked to an Fc region.
[0192] Human antibodies may be derived from phage-display technology or from
transgenic
mice that express human immunoglobulin genes. The human antibody may be
generated as a
result of a human in vivo immune response and isolated. See, for example,
Funaro etal.,
BMC Biotechnology, 2008(8):85. Therefore, the antibody may be a product of the
human
and not animal repertoire. Because it is of human origin, the risks of
reactivity against self-
antigens may be minimized. Alternatively, standard yeast display libraries and
display
technologies may be used to select and isolate human anti-UCH-L1 antibodies.
For example,
libraries of naive human single chain variable fragments (scFv) may be used to
select human
anti-UCH-Li antibodies. Transgenic animals may be used to express human
antibodies.
[0193] Humanized antibodies may be antibody molecules from non-human
species antibody
that binds the desired antigen having one or more complementarity determining
regions
(CDRs) from the non-human species and framework regions from a human
immunoglobulin
molecule.
[0194] The antibody is distinguishable from known antibodies in that it
possesses different
biological function(s) than those known in the art.
(1) Epitope
[0195] The antibody may immunospecifically bind to UCH-Li (SEQ ID NO: I), a
fragment
thereof, or a variant thereof. The antibody may immunospecifically recognize
and bind at
least three amino acids, at least four amino acids, at least five amino acids,
at least six amino
acids, at least seven amino acids, at least eight amino acids, at least nine
amino acids, or at
least ten amino acids within an epitope region. The antibody may
immunospecifically
recognize and bind to an epitope that has at least three contiguous amino
acids, at least four
contiguous amino acids, at least five contiguous amino acids, at least six
contiguous amino
69

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
acids, at least seven contiguous amino acids, at least eight contiguous amino
acids, at least
nine contiguous amino acids, or at least ten contiguous amino acids of an
epitope region.
c) Antibody Preparation/Production
[0196] Antibodies may be prepared by any of a variety of techniques,
including those well
known to those skilled in the art. In general, antibodies can be produced by
cell culture
techniques, including the generation of monoclonal antibodies via conventional
techniques,
or via transfection of antibody genes, heavy chains, and/or light chains into
suitable bacterial
or mammalian cell hosts, in order to allow for the production of antibodies,
wherein the
antibodies may be recombinant. The various forms of the term "transfection"
are intended to
encompass a wide variety of techniques commonly used for the introduction of
exogenous
DNA into a prokaryotic or eukaryotic host cell, e.g., electroporation, calcium-
phosphate
precipitation, DEAE-dextran transfection and the like. Although it is possible
to express the
antibodies in either prokaryotic or eukaryotic host cells, expression of
antibodies in
eukaryotic cells is preferable, and most preferable in mammalian host cells,
because such
eukaryotic cells (and in particular mammalian cells) are more likely than
prokaryotic cells to
assemble and secrete a properly folded and immunologically active antibody.
[0197] Exemplary mammalian host cells for expressing the recombinant
antibodies include
Chinese Hamster Ovary (CHO cells) (including dhfr-CHO cells, described in
Urlaub and
Chasin, Proc. Natl. Acad. Sci. USA, 77: 4216-4220 (1980)), used with a DHFR
selectable
marker, e.g., as described in Kaufman and Sharp, J. Mol. Biol., 159: 601-621
(1982), NSO
myeloma cells, COS cells, and SP2 cells. When recombinant expression vectors
encoding
antibody genes are introduced into mammalian host cells, the antibodies are
produced by
culturing the host cells for a period of time sufficient to allow for
expression of the antibody
in the host cells or, more preferably, secretion of the antibody into the
culture medium in
which the host cells are grown. Antibodies can be recovered from the culture
medium using
standard protein purification methods.
[0198] Host cells can also be used to produce functional antibody
fragments, such as Fab
fragments or scFv molecules. It will be understood that variations on the
above procedure
may be performed. For example, it may be desirable to transfect a host cell
with DNA

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
encoding functional fragments of either the light chain and/or the heavy chain
of an antibody.
Recombinant DNA technology may also be used to remove some, or all, of the DNA
encoding either or both of the light and heavy chains that is not necessary
for binding to the
antigens of interest. The molecules expressed from such truncated DNA
molecules are also
encompassed by the antibodies, In addition, bifunctional antibodies may be
produced in
which one heavy and one light chain are an antibody (i.e., binds human UCH-L1)
and the
other heavy and light chain are specific for an antigen other than human UCH-
L1 by
crosslinking an antibody to a second antibody by standard chemical
crosslinking methods.
[0199] In a preferred system for recombinant expression of an antibody, or
antigen-binding
portion thereof, a recombinant expression vector encoding both the antibody
heavy chain and
the antibody light chain is introduced into dhfr-CHO cells by calcium
phosphate-mediated
transfection. Within the recombinant expression vector, the antibody heavy and
light chain
genes are each operatively linked to CMV enhancer/AdMLP promoter regulatory
elements to
drive high levels of transcription of the genes. The recombinant expression
vector also
carries a DHFR gene, which allows for selection of CHO cells that have been
transfected
with the vector using methotrexate selection/amplification. The selected
transformant host
cells are cultured to allow for expression of the antibody heavy and light
chains and intact
antibody is recovered from the culture medium. Standard molecular biology
techniques are
used to prepare the recombinant expression vector, transfect the host cells,
select for
transformants, culture the host cells, and recover the antibody from the
culture medium. Still
further, the method of synthesizing a recombinant antibody may be by culturing
a host cell in
a suitable culture medium until a recombinant antibody is synthesized. The
method can
further comprise isolating the recombinant antibody from the culture medium.
[0200] Methods of preparing monoclonal antibodies involve the preparation
of immortal cell
lines capable of producing antibodies having the desired specificity. Such
cell lines may be
produced from spleen cells obtained from an immunized animal. The animal may
be
immunized with UCH-L1 or a fragment and/or variant thereof The peptide used to
immunize the animal may comprise amino acids encoding human Fc, for example
the
fragment crystallizable region or tail region of human antibody. The spleen
cells may then
be immortalized by, for example, fusion with a myeloma cell fusion partner. A
variety of
Ti

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
fusion techniques may be employed. For example, the spleen cells and myeloma
cells may
be combined with a nonionic detergent for a few minutes and then plated at low
density on a
selective medium that supports that growth of hybrid cells, but not myeloma
cells. One such
technique uses hypoxanthine, aminopterin, thymidine (HAT) selection. Another
technique
includes electrofusion. After a sufficient time, usually about 1 to 2 weeks,
colonies of
hybrids are observed. Single colonies are selected and their culture
supernatants tested for
binding activity against the polypeptide. Hybridomas having high reactivity
and specificity
may be used.
[0201] Monoclonal antibodies may be isolated from the supernatants of growing
hybridoma
colonies. In addition, various techniques may be employed to enhance the
yield, such as
injection of the hybridoma cell line into the peritoneal cavity of a suitable
vertebrate host,
such as a mouse. Monoclonal antibodies may then be harvested from the ascites
fluid or the
blood. Contaminants may be removed from the antibodies by conventional
techniques, such
as chromatography, gel filtration, precipitation, and extraction. Affinity
chromatography is
an example of a method that can be used in a process to purify the antibodies.
[0202] The proteolytic enzyme papain preferentially cleaves IgG molecules
to yield several
fragments, two of which (the F(ab) fragments) each comprise a covalent
heterodimer that
includes an intact antigen-binding site. The enzyme pepsin is able to cleave
IgG molecules
to provide several fragments, including the F(ab')2 fragment, which comprises
both antigen-
binding sites.
[0203] The Fv fragment can be produced by preferential proteolytic cleavage
of an IgM, and
on rare occasions IgG or IgA immunoglobulin molecules. The Fv fragment may be
derived
using recombinant techniques. The Fv fragment includes a non-covalent VH::VL
heterodimer including an antigen-binding site that retains much of the antigen
recognition
and binding capabilities of the native antibody molecule.
[0204] The antibody, antibody fragment, or derivative may comprise a heavy
chain and a light
chain complementarity determining region ("CDR") set, respectively interposed
between a
heavy chain and a light chain framework ("FR") set which provide support to
the CDRs and
define the spatial relationship of the CDRs relative to each other. The CDR
set may contain
three hypervariable regions of a heavy or light chain V region.
72

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0205] Other suitable methods of producing or isolating antibodies of the
requisite specificity
can be used, including, but not limited to, methods that select recombinant
antibody from a
peptide or protein library (e.g., but not limited to, a bacteriophage,
ribosome, oligonucleotide,
RNA, cDNA, yeast or the like, display library); e.g., as available from
various commercial
vendors such as Cambridge Antibody Technologies (Cambridgeshire, UK),
MorphoSys
(Martinsreid/Planegg, Del.), Biovation (Aberdeen, Scotland, UK) BioInvent
(Lund, Sweden),
using methods known in the art. See U.S. Patent Nos. 4,704,692; 5,723,323;
5,763,192;
5,814,476; 5,817,483; 5,824,514; 5,976,862. Alternative methods rely upon
immunization of
transgenic animals (e.g., SCID mice, Nguyen etal. (1997)Microbio/. Iminunol.
41:901-907;
Sandhu et al. (1996) Crit. Rev. BlotechnoL 16:95-118; Eren et al. (1998)
ImmunoL 93:154-
161) that are capable of producing a repertoire of human antibodies, as known
in the art
and/or as described herein. Such techniques, include, but are not limited to,
ribosome display
(Hanes et al. (1997) Proc. Natl. Acad. Sci. USA, 94:4937-4942; Hanes et al.
(1998) Proc.
Natl. Acad. Sci. USA, 95:14130-14135); single cell antibody producing
technologies (e.g.,
selected lymphocyte antibody method ("SLAM") (U.S. Patent No. 5,627,052, Wen
et al.
(1987)J. ImmunoL 17:887-892; lBabcooketai. (1996) Proc. Natl. Acad Sci. USA
93:7843-
7848); gel microdroplet and flow cytometry (Powell et al. (1990) Biotechnol.
8:333-337;
One Cell Systems, (Cambridge, Mass).; Gray etal. (1995) J. Imm. Meth. 182:155-
163;
Kenny et al. (1995) Bio/Technol. 13:787-790); B-cell selection (Steenbakkers
etal. (1994)
Molec. Biol. Reports 19:125-134 (1994)).
[0206] An affinity matured antibody may be produced by any one of a number of
procedures
that are known in the art. For example, see Marks et al ., BioTechnology, 10:
779-783 (1992)
describes affinity maturation by VH and VL domain shuffling. Random
mutagenesis of CDR
and/or framework residues is described by Barbas et al., Proc. Nat. Acad. Sci.
USA, 91:
3809-3813 (1994); Schier etal., Gene, 169: 147-155 (1995); Yelton etal., J.
Immunol., 155:
1994-2004 (1995); Jackson et al., J. Immunol., 154(7): 3310-3319 (1995);
Hawkins eta!, J.
Mot Biol., 226: 889-896 (1992). Selective mutation at selective mutagenesis
positions and at
contact or hypermutation positions with an activity enhancing amino acid
residue is
described in U.S. Patent No. 6,914,128 Bl.
73

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0207] Antibody variants can also be prepared using delivering a
polynucleotide encoding an
antibody to a suitable host such as to provide transgenic animals or mammals,
such as goats,
cows, horses, sheep, and the like, that produce such antibodies in their milk.
These methods
are known in the art and are described for example in U.S. Patent Nos.
5,827,690; 5,849,992;
4,873,316; 5,849,992; 5,994,616; 5,565,362; and 5,304,489.
[0208] Antibody variants also can be prepared by delivering a
polynucleotide to provide
transgenic plants and cultured plant cells (e.g., but not limited to tobacco,
maize, and
duckweed) that produce such antibodies, specified portions or variants in the
plant parts or in
cells cultured therefrom. For example, Cramer et at. (1999) Curr. Top.
Microbiol. Imrnunol.
240:95-118 and references cited therein, describe the production of transgenic
tobacco leaves
expressing large amounts of recombinant proteins, e.g., using an inducible
promoter.
Transgenic maize have been used to express mammalian proteins at commercial
production
levels, with biological activities equivalent to those produced in other
recombinant systems
or purified from natural sources. See, e.g., Hood et at., Adv. Exp. Med. Biol.
(1999) 464:127-
147 and references cited therein. Antibody variants have also been produced in
large
amounts from transgenic plant seeds including antibody fragments, such as
single chain
antibodies (scFv's), including tobacco seeds and potato tubers. See, e.g.,
Conrad et al. (1998)
Plant Mol. Biol. 38:101-109 and reference cited therein. Thus, antibodies can
also be
produced using transgenic plants, according to known methods.
[0209] Antibody derivatives can be produced, for example, by adding
exogenous sequences to
modify immunogenicity or reduce, enhance or modify binding, affinity, on-rate,
off-rate,
avidity, specificity, half-life, or any other suitable characteristic.
Generally, part or all of the
non-human or human CDR sequences are maintained while the non-human sequences
of the
variable and constant regions are replaced with human or other amino acids.
[0210] Small antibody fragments may be diabodies having two antigen-binding
sites, wherein
fragments comprise a heavy chain variable domain (VH) connected to a light
chain variable
domain (VL) in the same polypeptide chain (VIA VL). See for example, EP
404,097; WO
93/11161; and Hollinger etal., (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448.
By using a
linker that is too short to allow pairing between the two domains on the same
chain, the
domains are forced to pair with the complementary domains of another chain and
create two
74

antigen-binding sites. See also, U.S. Patent No. 6,632,926 to Chen et al.
which
discloses antibody variants that have one or more
amino acids inserted into a hypervariable region of the parent antibody and a
binding affinity
for a target antigen which is at least about two fold stronger than the
binding affinity of the
parent antibody for the antigen.
[0211] The antibody may be a linear antibody. The procedure for making a
linear antibody is
known in the art and described in Zapata etal., (1995) Protein Eng. 8(10):1057-
1062.
Briefly, these antibodies comprise a pair of tandem Fd segments (VH-CH1-VH-
CH1) which
form a pair of antigen binding regions. Linear antibodies can be bispecific or
monospecific.
[0212] The antibodies may be recovered and purified from recombinant cell
cultures by
known methods including, but not limited to, protein A purification, ammonium
sulfate or
ethanol precipitation, acid extraction, anion or cation exchange
chromatography,
phosphocellulose chromatography, hydrophobic interaction chromatography,
affinity
chromatography, hydroxylapatite chromatography and lectin chromatography. High
performance liquid chromatography ("HPLC") can also be used for purification.
[0213] It may be useful to detectably label the antibody. Methods for
conjugating antibodies
to these agents are known in the art. For the purpose of illustration only,
antibodies can be
labeled with a detectable moiety such as a radioactive atom, a chromophore, a
fluorophore,
or the like. Such labeled antibodies can be used for diagnostic techniques,
either in vivo, or
in an isolated test sample. They can be linked to a cytokine, to a ligand, to
another antibody.
Suitable agents for coupling to antibodies to achieve an anti-tumor effect
include cytoldnes,
such as interleukin 2 (IL-2) and Tumor Necrosis Factor (TNF);
photosensitizers, for use in
photodynamic therapy, including aluminum (1II) phthalocyanine tetrasulfonate,
hematoporphyrin, and phthalocyanine; radionuclides, such as iodine-131 (1311),
yttrium-90
(90Y), bismuth-212 (212Bi), bismuth-213 (213Bi), technetium-99m (99mTc),
rhenium-186
(186Re), and rhenium-188 (188Re); antibiotics, such as doxorubicin,
adriamycin,
daunorubicin, methotrexate, daunomycin, neocarzinostatin, and carboplatin;
bacterial, plant,
and other toxins, such as diphtheria toxin, pseudomonas exotoxin A,
staphylococcal
enterotoxin A, abrin-A toxin, ricin A (deglycosylated ricin A and native ricin
A), TGF-alpha
toxin, cytotoxin from chinese cobra (naja naja atra), and gelonin (a plant
toxin); ribosome
Date Recue/Date Received 2023-06-29

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
inactivating proteins from plants, bacteria and fungi, such as restrictocin (a
ribosome
inactivating protein produced by Aspergillus restrictus), saporin (a ribosome
inactivating
protein from Saponaria officinalis), and RNase; tyrosine kinase inhibitors;
ly207702 (a
difluorinated purine nucleoside); liposomes containing anti cystic agents
(e.g., antisense
oligonucleotides, plasmids which encode for toxins, methotrexate, etc.); and
other antibodies
or antibody fragments, such as F(ab).
[0214] Antibody production via the use of hybridoma technology, the
selected lymphocyte
antibody method (SLAM), transgenic animals, and recombinant antibody libraries
is
described in more detail below.
(1) Anti-UCH-L1 Monoclonal Antibodies Using Hybridoma Technology
[0215] Monoclonal antibodies can be prepared using a wide variety of
techniques known in
the art including the use of hybridoma, recombinant, and phage display
technologies, or a
combination thereof. For example, monoclonal antibodies can be produced using
hybridoma
techniques including those known in the art and taught, for example, in Harlow
et al.,
Antibodies: A Laboratory Manual, second edition, (Cold Spring Harbor
Laboratory Press,
Cold Spring Harbor, 1988); Hammerling, et al., In Monoclonal Antibodies and T-
Cell
Hybridomas, (Elsevier, N.Y., 1981). It is also noted that the term "monoclonal
antibody" as
used herein is not limited to antibodies produced through hybridoma
technology. The term
"monoclonal antibody" refers to an antibody that is derived from a single
clone, including
any eukaryotic, prokaryotic, or phage clone, and not the method by which it is
produced.
[0216] Methods of generating monoclonal antibodies as well as antibodies
produced by the
method may comprise culturing a hybridoma cell secreting an antibody of the
invention
wherein, preferably, the hybridoma is generated by fusing splenocytes isolated
from an
animal, e.g., a rat or a mouse, immunized with UCH-L1 with myeloma cells and
then
screening the hybridomas resulting from the fusion for hybridoma clones that
secrete an
antibody able to bind a polypeptide of the invention. Briefly, rats can be
immunized with a
UCH-L1 antigen. In a preferred embodiment, the UCH-L1 antigen is administered
with an
adjuvant to stimulate the immune response. Such adjuvants include complete or
incomplete
Freund's adjuvant, RIM (muramyl dipeptides) or ISCOM (immunostimulating
complexes).
Such adjuvants may protect the polypeptide from rapid dispersal by
sequestering it in a local
76

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
deposit, or they may contain substances that stimulate the host to secrete
factors that are
chemotactic for macrophages and other components of the immune system.
Preferably, if a
polypeptide is being administered, the immunization schedule will involve two
or more
administrations of the polypeptide, spread out over several weeks; however, a
single
administration of the polypeptide may also be used.
[0217] After immunization of an animal with a UCH-L1 antigen, antibodies
and/or antibody-
producing cells may be obtained from the animal. An anti-UCH-L1 antibody-
containing
serum is obtained from the animal by bleeding or sacrificing the animal. The
serum may be
used as it is obtained from the animal, an immunoglobulin fraction may be
obtained from the
serum, or the anti-UCH-L1 antibodies may be purified from the serum. Serum or
immunoglobulins obtained in this manner are polyclonal, thus having a
heterogeneous array
of properties.
[0218] Once an immune response is detected, e.g., antibodies specific for
the antigen UCH-
Li are detected in the rat serum, the rat spleen is harvested and splenocytes
isolated. The
splenocytes are then fused by well-known techniques to any suitable myeloma
cells, for
example, cells from cell line SP20 available from the American Type Culture
Collection
(ATCC, Manassas, Va., US). Hybridomas are selected and cloned by limited
dilution. The
hybridoma clones are then assayed by methods known in the art for cells that
secrete
antibodies capable of binding UCH-Li. Ascites fluid, which generally contains
high levels
of antibodies, can be generated by immunizing rats with positive hybridoma
clones.
[0219] In another embodiment, antibody-producing immortalized hybridomas
may be
prepared from the immunized animal. After immunization, the animal is
sacrificed and the
splenic B cells are fused to immortalized myeloma cells as is well known in
the art See,
e.g., Harlow and Lane, supra. In a preferred embodiment, the myeloma cells do
not secrete
immunoglobulin polypeptides (a non-secretory cell line). After fusion and
antibiotic
selection, the hybridomas are screened using UCH-L1, or a portion thereof, or
a cell
expressing UCH-Li. In a preferred embodiment, the initial screening is
performed using an
enzyme-linked immunosorbent assay (ELISA) or a radioimmunoassay (RIA),
preferably an
ELISA. An example of ELISA screening is provided in PCT Publication No. WO
00/37504.
77

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0220] Anti-UCH-L1 antibody-producing hybridomas are selected, cloned, and
further
screened for desirable characteristics, including robust hybridoma growth,
high antibody
production, and desirable antibody characteristics. Hybridomas may be cultured
and
expanded in vivo in syngeneic animals, in animals that lack an immune system,
e.g., nude
mice, or in cell culture in vitro. Methods of selecting, cloning and expanding
hybridomas are
well known to those of ordinary skill in the art.
[0221] In a preferred embodiment, hybridomas are rat hybridomas. In another
embodiment,
hybridomas are produced in a non-human, non-rat species such as mice, sheep,
pigs, goats,
cattle, or horses. In yet another preferred embodiment, the hybridomas are
human
hybridomas, in which a human non-secretory myeloma is fused with a human cell
expressing
an anti-UCH-L1 antibody.
[0222] Antibody fragments that recognize specific epitopes may be generated
by known
techniques. For example, Fab and F(ab')2 fragments of the invention may be
produced by
proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain
(to
produce two identical Fab fragments) or pepsin (to produce an F(ab')2
fragment). A F(ab')2
fragment of an IgG molecule retains the two antigen-binding sites of the
larger ("parent")
IgG molecule, including both light chains (containing the variable light chain
and constant
light chain regions), the CHI domains of the heavy chains, and a disulfide-
forming hinge
region of the parent IgG molecule. Accordingly, an F(ab')2 fragment is still
capable of
crosslinking antigen molecules like the parent IgG molecule.
(2) Anti-UCH-L1 Monoclonal Antibodies Using SLAM
[0223] In another aspect of the invention, recombinant antibodies are
generated from single,
isolated lymphocytes using a procedure referred to in the art as the selected
lymphocyte
antibody method (SLAM), as described in U.S. Patent No. 5,627,052; PCT
Publication No.
WO 92/02551; and Babcook et al., Proc. Natl. Acad. Sci. USA, 93: 7843-7848
(1996). In this
method, single cells secreting antibodies of interest, e.g., lymphocytes
derived from any one
of the immunized animals are screened using an antigen-specific hemolytic
plaque assay,
wherein the antigen UCH-L1, a subunit of UCH-L1, or a fragment thereof, is
coupled to
sheep red blood cells using a linker, such as biotin, and used to identify
single cells that
secrete antibodies with specificity for UCH-Li. Following identification of
antibody-
78

secreting cells of interest, heavy- and light-chain variable region cDNAs are
rescued from the
cells by reverse transcriptase-PCR (RT-PCR) and these variable regions can
then be
expressed, in the context of appropriate immunoglobulin constant regions
(e.g., human
constant regions), in mammalian host cells, such as COS or CHO cells. The host
cells
transfected with the amplified immunoglobulin sequences, derived from in vivo
selected
lymphocytes, can then undergo further analysis and selection in vitro, for
example, by
panning the transfected cells to isolate cells expressing antibodies to UCH-
LI. The amplified
immunoglobulin sequences further can be manipulated in vitro, such as by in
vitro affinity
maturation method. See, for example, PCT Publication No. WO 97/29131 and PCT
Publication No. WO 00/56772.
(3) Anti-UCH-L11 Monoclonal Antibodies Using Transgenic Animals
[0224] In another embodiment of the invention, antibodies are produced by
immunizing a
non-human animal comprising some, or all, of the human immunoglobulin locus
with a
UCH-L1 antigen. In an embodiment, the non-human animal is a XENOMOUSE
transgenic
mouse, an engineered mouse strain that comprises large fragments of the human
immunoglobulin loci and is deficient in mouse antibody production. See, e.g.,
Green et al.,
Nature Genetics, 7: 13-21 (1994) and U.S. Patent Nos. 5,916,771; 5,939,598;
5,985,615;
5,998,209; 6,075,181; 6,091,001; 6,114,598; and 6,130,364. See also PCT
Publication Nos.
WO 91/10741; WO 94/02602; WO 96/34096; WO 96/33735; WO 98/16654; WO 98/24893;
WO 98/50433; WO 99/45031; WO 99/53049; WO 00/09560; and WO 00/37504. The
XENOMOUSE transgenic mouse produces an adult-like human repertoire of fully
human
antibodies, and generates antigen-specific human monoclonal antibodies. The
XENOMOUSE 0 transgenic mouse contains approximately 80% of the human antibody
repertoire through introduction of megabase sized, germline configuration YAC
fragments of
the human heavy chain loci and x light chain loci. See Mendez et al., Nature
Genetics, 15:
146-156 (1997), Green and Jakobovits, J. Exp. Med., 188: 483-495 (1998).
79
Date Recue/Date Received 2023-06-29

(4) Anti-UCH-L1 Monoclonal Antibodies Using Recombinant Antibody
Libraries
[0225] In vitro methods also can be used to make the antibodies of the
invention, wherein an
antibody library is screened to identify an antibody having the desired UCH-L1
-binding
specificity. Methods for such screening of recombinant antibody libraries are
well known in
the art and include methods described in, for example, U.S. Patent No.
5,223,409 (Ladner et
al.); PCT Publication No. WO 92/18619 (Kang et al.); PCT Publication No. WO
91/17271
(Dower et al.); PCT Publication No. WO 92/20791 (Winter etal.); PCT
Publication No. WO
92/15679 (Marldand et al.); PCT Publication No. WO 93/01288 (Breitling et
al.); PCT
Publication No. WO 92/01047 (McCafferty etal.); PCT Publication No. WO
92/09690
(Garrard etal.); Fuchs etal., Bio/Technology, 9: 1369-1372 (1991); Hay et al.,
Hum.
Antibod. Hybridomas, 3: 81-85 (1992); Huse etal., Science, 246: 1275-1281
(1989);
McCafferty etal., Nature, 348: 552-554 (1990); Griffiths etal., EMBO J., 12:
725-734
(1993); Hawkins et al., J. Mol. Biol., 226: 889-896 (1992); Clackson et al.,
Nature, 352: 624-
628 (1991); Gram etal., Proc. Natl. Acad. Sci. USA, 89: 3576-3580 (1992);
Garrard et al.,
Bio/Technology, 9: 1373-1377 (1991); Hoogenboom etal., Nucl. Acids Res., 19:
4133-4137
(1991); Barbas etal., Proc. Natl. Acad. Sc!. USA, 88: 7978-7982 (1991); U.S.
Patent
Application Publication No. 2003/0186374; and PCT Publication No. WO 97/29131.
[0226] The recombinant antibody library may be from a subject immunized with
UCH-L1, or
a portion of UCH-L I. Alternatively, the recombinant antibody library may be
from a naive
subject, i.e., one who has not been immunized with UCH-L1, such as a human
antibody
library from a human subject who has not been immunized with human UCH-Li.
Antibodies
of the invention are selected by screening the recombinant antibody library
with the peptide
comprising human UCH-L1 to thereby select those antibodies that recognize UCH-
L1.
Methods for conducting such screening and selection are well known in the art,
such as
described in the references in the preceding paragraph. To select antibodies
of the invention
having particular binding affinities for UCH-L1, such as those that dissociate
from human
UCH-L1 with a particular Koff rate constant, the art-known method of surface
plasmon
resonance can be used to select antibodies having the desired Koff rate
constant. To select
Date Recue/Date Received 2023-06-29

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
antibodies of the invention having a particular neutralizing activity for hUCH-
L1, such as
those with a particular IC50, standard methods known in the art for assessing
the inhibition of
UCH-L1 activity may be used.
[0227] In one aspect, the invention pertains to an isolated antibody, or an
antigen-binding
portion thereof, that binds human UCH-Li. Preferably, the antibody is a
neutralizing
antibody. In various embodiments, the antibody is a recombinant antibody or a
monoclonal
antibody.
[0228] For example, antibodies can also be generated using various phage
display methods
known in the art. In phage display methods, functional antibody domains are
displayed on the
surface of phage particles which carry the polynucleotide sequences encoding
them. Such
phage can be utilized to display antigen-binding domains expressed from a
repertoire or
combinatorial antibody library (e.g., human or murine). Phage expressing an
antigen binding
domain that binds the antigen of interest can be selected or identified with
antigen, e.g., using
labeled antigen or antigen bound or captured to a solid surface or bead. Phage
used in these
methods are typically filamentous phage including fd and M13 binding domains
expressed
from phage with Fab, Fv, or disulfide stabilized Fv antibody domains
recombinantly fused to
either the phage gene HI or gene VIII protein. Examples of phage display
methods that can
be used to make the antibodies include those disclosed in Brinkmann et at., I
Alumna
Methods, 182: 41-50 (1995); Ames etal., J. Immunol. Methods, 184:177-186
(1995);
Kettleborough et al., Eur. I Immunol., 24: 952-958 (1994); Persic et at.,
Gene, 187: 9-18
(1997); Burton et at., Advances in Immunology, 57: 191-280 (1994); PCT
Publication No.
WO 92/01047; PCT Publication Nos. WO 90/02809; WO 91/10737; WO 92/01047; WO
92/18619; WO 93/11236; WO 95/15982; WO 95/20401; and U.S. Patent Nos.
5,698,426;
5,223,409; 5,403,484; 5,580,717; 5,427,908; 5,750,753; 5,821,047; 5,571,698;
5,427,908;
5,516,637; 5,780,225; 5,658,727; 5,733,743; and 5,969,108.
[0229] As described in the above references, after phage selection, the
antibody coding
regions from the phage can be isolated and used to generate whole antibodies
including
human antibodies or any other desired antigen binding fragment, and expressed
in any
desired host, including mammalian cells, insect cells, plant cells, yeast, and
bacteria, e.g., as
described in detail below. For example, techniques to recombinantly produce
Fab, Fab', and
81

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
F(a1:02 fragments can also be employed using methods known in the art such as
those
disclosed in PCT publication No. WO 92/22324; Mullinax et al., BioTechniques,
12(6): 864-
869 (1992); Sawai et al., Am. J. Reprod Immunol., 34: 26-34 (1995); and Better
etal.,
Science, 240: 1041-1043 (1988). Examples of techniques which can be used to
produce
single-chain Fvs and antibodies include those described in U.S. Patent Nos.
4,946,778 and
5,258,498; Huston et al., Methods in Enzymology, 203: 46-88 (1991); Shu et
al., Proc. Natl.
Acad. S'ci. USA, 90: 7995-7999 (1993); and Skerra etal., Science, 240: 1038-
1041 (1988).
[0230] Alternative to screening of recombinant antibody libraries by phage
display, other
methodologies known in the art for screening large combinatorial libraries can
be applied to
the identification of antibodies of the invention. One type of alternative
expression system is
one in which the recombinant antibody library is expressed as RNA-protein
fusions, as
described in PCT Publication No. WO 98/31700 (Szostak and Roberts), and in
Roberts and
Szostak, Proc. Natl. Acad. Sci. USA, 94: 12297-12302 (1997). In this system, a
covalent
fusion is created between an mRNA and the peptide or protein that it encodes
by in vitro
translation of synthetic mRNAs that carry puromycin, a peptidyl acceptor
antibiotic, at their
3' end. Thus, a specific mRNA can be enriched from a complex mixture of mRNAs
(e.g., a
combinatorial library) based on the properties of the encoded peptide or
protein, e.g.,
antibody, or portion thereof, such as binding of the antibody, or portion
thereof, to the dual
specificity antigen. Nucleic acid sequences encoding antibodies, or portions
thereof,
recovered from screening of such libraries can be expressed by recombinant
means as
described above (e.g., in mammalian host cells) and, moreover, can be
subjected to further
affinity maturation by either additional rounds of screening of mRNA-peptide
fusions in
which mutations have been introduced into the originally selected sequence(s),
or by other
methods for affinity maturation in vitro of recombinant antibodies, as
described above. A
preferred example of this methodology is PROfusion display technology.
[0231] In another approach, the antibodies can also be generated using
yeast display methods
known in the art. In yeast display methods, genetic methods are used to tether
antibody
domains to the yeast cell wall and display them on the surface of yeast. In
particular, such
yeast can be utilized to display antigen-binding domains expressed from a
repertoire or
combinatorial antibody library (e.g., human or murine). Examples of yeast
display methods
82

that can be used to make the antibodies include those disclosed in U.S. Patent
No. 6,699,658
(Wittrup et al.).
d) Production of Recombinant UCH-L1 Antibodies
[0232] Antibodies may be produced by any of a number of techniques known in
the art. For
example, expression from host cells, wherein expression vector(s) encoding the
heavy and
light chains is (are) transfected into a host cell by standard techniques. The
various forms of
the term "transfection" are intended to encompass a wide variety of techniques
commonly
used for the introduction of exogenous DNA into a prokaryotic or eukaryotic
host cell, e.g.,
electroporation, calcium-phosphate precipitation, DEAE-dextran transfection,
and the like.
Although it is possible to express the antibodies of the invention in either
prokaryotic or
eukaryotic host cells, expression of antibodies in eukaryotic cells is
preferable, and most
preferable in mammalian host cells, because such eukaryotic cells (and in
particular
mammalian cells) are more likely than prokaryotic cells to assemble and
secrete a properly
folded and immunologically active antibody.
[0233] Exemplary mammalian host cells for expressing the recombinant
antibodies of the
invention include Chinese Hamster Ovary (CEO cells) (including dhfr-CHO cells,
described
in Urlaub and Chasin, Proc. Natl. Accra/. Sri. USA, 77: 4216-4220 (1980), used
with a DHFR
selectable marker, e.g., as described in Kaufman and Sharp, Mol. Biol., 159:
601-621
(1982), NSO myeloma cells, COS cells, and SP2 cells. When recombinant
expression vectors
encoding antibody genes are introduced into mammalian host cells, the
antibodies are
produced by culturing the host cells for a period of time sufficient to allow
for expression of
the antibody in the host cells or, more preferably, secretion of the antibody
into the culture
medium in which the host cells are grown. Antibodies can be recovered from the
culture
medium using standard protein purification methods.
[0234] Host cells can also be used to produce functional antibody fragments,
such as Fab
fragments or scFv molecules. It will be understood that variations on the
above procedure
may be performed. For example, it may be desirable to transfect a host cell
with DNA
encoding functional fragments of either the light chain and/or the heavy chain
of an antibody
of this invention. Recombinant DNA technology may also be used to remove some,
or all, of
83
Date Recue/Date Received 2023-06-29

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
the DNA encoding either or both of the light and heavy chains that is not
necessary for
binding to the antigens of interest. The molecules expressed from such
truncated DNA
molecules are also encompassed by the antibodies of the invention. In
addition, bifunctional
antibodies may be produced in which one heavy and one light chain are an
antibody of the
invention (i.e., binds human UCH-L1) and the other heavy and light chain are
specific for an
antigen other than human UCH-L1 by crosslinking an antibody of the invention
to a second
antibody by standard chemical crosslinking methods.
[0235] In a preferred system for recombinant expression of an antibody, or
antigen-binding
portion thereof, of the invention, a recombinant expression vector encoding
both the antibody
heavy chain and the antibody light chain is introduced into dhfr-CHO cells by
calcium
phosphate-mediated transfection. Within the recombinant expression vector, the
antibody
heavy and light chain genes are each operatively linked to CMV enhancer/AdMLP
promoter
regulatory elements to drive high levels of transcription of the genes. The
recombinant
expression vector also carries a DHFR gene, which allows for selection of CHO
cells that
have been transfected with the vector using methotrexate
selection/amplification. The
selected transformant host cells are cultured to allow for expression of the
antibody heavy
and light chains and intact antibody is recovered from the culture medium.
Standard
molecular biology techniques are used to prepare the recombinant expression
vector,
transfect the host cells, select for transformants, culture the host cells,
and recover the
antibody from the culture medium. Still further, the invention provides a
method of
synthesizing a recombinant antibody of the invention by culturing a host cell
of the invention
in a suitable culture medium until a recombinant antibody of the invention is
synthesized.
The method can further comprise isolating the recombinant antibody from the
culture
medium.
(1) Humanized Antibody
[0236] The humanized antibody may be an antibody or a variant, derivative,
analog or portion
thereof which immunospecifically binds to an antigen of interest and which
comprises a
framework (FR) region having substantially the amino acid sequence of a human
antibody
and a complementary determining region (CDR) having substantially the amino
acid
sequence of a non-human antibody. The humanized antibody may be from a non-
human
84

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
species antibody that binds the desired antigen having one or more
complementarity
determining regions (CDRs) from the non-human species and framework regions
from a
human immunoglobulin molecule.
[0237] As used herein, the term "substantially" in the context of a CDR
refers to a CDR
having an amino acid sequence at least 90%, at least 95%, at least 98% or at
least 99%
identical to the amino acid sequence of a non-human antibody CDR. A humanized
antibody
comprises substantially all of at least one, and typically two, variable
domains (Fab, Fab',
F(ab)2, FabC, Fv) in which all or substantially all of the CDR regions
correspond to those of
a non-human immunoglobulin (i.e., donor antibody) and all or substantially all
of the
framework regions are those of a human immunoglobulin consensus sequence.
According to
one aspect, a humanized antibody also comprises at least a portion of an
immunoglobulin
constant region (Fc), typically that of a human immunoglobulin. In some
embodiments, a
humanized antibody contains both the light chain as well as at least the
variable domain of a
heavy chain. The antibody also may include the CH1, hinge, CH2, CH3, and CH4
regions of
the heavy chain. In some embodiments, a humanized antibody only contains a
humanized
light chain. In some embodiments, a humanized antibody only contains a
humanized heavy
chain. In specific embodiments, a humanized antibody only contains a humanized
variable
domain of a light chain and/or of a heavy chain.
[0238] The humanized antibody can be selected from any class of
immunoglobulins,
including IgM, IgG, IgD, IgA and IgE, and any isotype, including without
limitation IgG 1,
IgG2, IgG3, and IgG4. The humanized antibody may comprise sequences from more
than
one class or isotype, and particular constant domains may be selected to
optimize desired
effector functions using techniques well-known in the art
[0239] The framework and CDR regions of a humanized antibody need not
correspond
precisely to the parental sequences, e.g., the donor antibody CDR or the
consensus
framework may be mutagenized by substitution, insertion and/or deletion of at
least one
amino acid residue so that the CDR or framework residue at that site does not
correspond to
either the donor antibody or the consensus framework. In one embodiment, such
mutations,
however, will not be extensive. Usually, at least 90%, at least 95%, at least
98%, or at least
99% of the humanized antibody residues will correspond to those of the
parental FR and

CDR sequences. As used herein, the term "consensus framework" refers to the
framework
region in the consensus immunoglobulin sequence. As used herein, the term
"consensus
immunoglobulin sequence" refers to the sequence formed from the most
frequently occurring
amino acids (or nucleotides) in a family of related immunoglobulin sequences
(See e.g.,
Winnaker, From Genes to Clones (Verlagsgesellschaft, Weinheim, Germany 1987)).
In a
family of immunoglobulins, each position in the consensus sequence is occupied
by the
amino acid occurring most frequently at that position in the family. If two
amino acids occur
equally frequently, either can be included in the consensus sequence.
[0240] The humanized antibody may be designed to minimize unwanted
immunological
response toward rodent anti-human antibodies, which limits the duration and
effectiveness of
therapeutic applications of those moieties in human recipients. The humanized
antibody may
have one or more amino acid residues introduced into it from a source that is
non-human.
These non-human residues are often referred to as "import" residues, which are
typically
taken from a variable domain. Humanization may be performed by substituting
hypervariable region sequences for the corresponding sequences of a human
antibody.
Accordingly, such "humanized" antibodies are chimeric antibodies wherein
substantially less
than an intact human variable domain has been substituted by the corresponding
sequence
from a non-human species. For example, see U.S. Patent No. 4,816,567.
The humanized antibody may be a human
antibody in which some hypervariable region residues, and possibly some FR
residues are
substituted by residues from analogous sites in rodent antibodies.
Humanization or
engineering of antibodies of the present disclosure can be performed using any
known
method, such as but not limited to those described in U.S. Patent Nos.
5,723,323; 5,976,862;
5,824,514; 5,817,483; 5,814,476; 5,763,192; 5,723,323; 5,766,886; 5,714,352;
6,204,023;
6,180,370; 5,693,762; 5,530,101; 5,585,089; 5,225,539; and 4,816,567.
[0241] The humanized antibody may retain high affinity for UCH-L1 and other
favorable
biological properties. The humanized antibody may be prepared by a process of
analysis of
the parental sequences and various conceptual humanized products using three-
dimensional
models of the parental and humanized sequences. Three-dimensional
immunoglobulin
models are commonly available. Computer programs are available that illustrate
and display
86
Date Recue/Date Received 2023-06-29

probable three-dimensional conformational structures of selected candidate
immunoglobulin
sequences. Inspection of these displays permits analysis of the likely role of
the residues in
the functioning of the candidate immunoglobulin sequence, i.e., the analysis
of residues that
influence the ability of the candidate immunoglobulin to bind its antigen. In
this way, FR
residues can be selected and combined from the recipient and import sequences
so that the
desired antibody characteristics, such as increased affinity for UCH-L1, is
achieved. In
general, the hypervariable region residues may be directly and most
substantially involved in
influencing antigen binding.
[0242] As an alternative to humanization, human antibodies (also referred
to herein as "fully
human antibodies") can be generated. For example, it is possible to isolate
human antibodies
from libraries via PROfusion and/or yeast related technologies. It is also
possible to produce
transgenic animals (e.g., mice that are capable, upon immunization, of
producing a full
repertoire of human antibodies in the absence of endogenous immunoglobulin
production.
For example, the homozygous deletion of the antibody heavy-chain joining
region (JH) gene
in chimeric and genii-line mutant mice results in complete inhibition of
endogenous antibody
production. Transfer of the human germ-line immunoglobulin gene array in such
germ-line
mutant mice will result in the production of human antibodies upon antigen
challenge. The
humanized or fully human antibodies may be prepared according to the methods
described in
U.S. Patent Nos. 5,770,429; 5,833,985; 5,837,243; 5,922,845; 6,017,517;
6,096,311;
6,111,166; 6,270,765; 6,303,755; 6,365,116; 6,410,690; 6,682,928; and
6,984,720.
e) Anti-UCH-Li antibodies
[0243] Anti-UCH-Li antibodies may be generated using the techniques described
above as
well as using routine techniques known in the art. In some embodiments, the
anti-UCH-L1
antibody may be an unconjugated UCH-L1 antibody, such as UCH-LI antibodies
available
from United State Biological (Catalog Number: 031320), Cell Signaling
Technology
(Catalog Number: 3524), Sigma-Aldrich (Catalog Number: HPA005993), Santa Cruz
Biotechnology, Inc. (Catalog Numbers: sc-58593 or sc-58594), R&D Systems
(Catalog
Number: MAB6007), Novus Biologicals (Catalog Number: NB600-1160), Biorbyt
(Catalog
87
Date Recue/Date Received 2023-06-29

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
Number: orb33715), Enzo Life Sciences, Inc. (Catalog Number: ADI-905-520-1),
Bio-Rad
(Catalog Number: VMA00004), BioVision (Catalog Number: 6130-50), Abcam
(Catalog
Numbers: ab75275 or ab104938), Invitrogen Antibodies (Catalog Numbers:
480012),
ThermoFisher Scientific (Catalog Numbers: MA1-46079, MA5-17235, MAI-90008, or
MA1-83428), EMD Millipore (Catalog Number: MABN48), or Sino Biological Inc.
(Catalog
Number: 50690-R011). The anti-UCH-Li antibody may be conjugated to a
fluorophore, such
as conjugated UCH-L1 antibodies available from BioVision (Catalog Number: 6960-
25) or
Aviva Systems Biology (Cat. Nos. OAAF01904-FITC).
13. Variations on Methods
[0244] The disclosed methods of determining the presence or amount of UCH-
L1 present in a
sample may be as described herein. The methods may also be adapted in view of
other
methods for analyzing analytes. Examples of well-known variations include, but
are not
limited to, immunoassay, such as sandwich immunoassay (e.g., monoclonal-
monoclonal
sandwich immunoassays, monoclonal-polyclonal sandwich immunoassays, including
enzyme
detection (enzyme immunoassay (EIA) or enzyme-linked immunosorbent assay
(ELISA),
competitive inhibition immunoassay (e.g., forward and reverse), enzyme
multiplied
immunoassay technique (EMIT), a competitive binding assay, bioluminescence
resonance
energy transfer (BRET), one-step antibody detection assay, homogeneous assay,
heterogeneous assay, capture on the fly assay, etc.
a) Immunoassays
[0245] The analyte of interest, and/or peptides of fragments thereof (e.g.,
UCH-L1, and/or
peptides or fragments thereof, i.e., UCH-L1 fragments), may be analyzed using
UCH-L1
antibodies in an immunoassay. The presence or amount of analyte (e.g., UCH-L1)
can be
determined using antibodies and detecting specific binding to the analyte
(e.g., UCH-L1). For
example, the antibody, or antibody fragment thereof, may specifically bind to
the analyte
(e.g., UCH-L1). If desired, one or more of the antibodies can be used in
combination with
one or more commercially available monoclonal/polyclonal antibodies. Such
antibodies are
available from companies such as R&D Systems, Inc. (Minneapolis, MN) and Enzo
Life
Sciences International, Inc. (Plymouth Meeting, PA).
88

[0246] The presence or amount of analyte (e.g., UCH-Li) present in a body
sample may be
readily determined using an immunoassay, such as sandwich immunoassay (e.g.,
monoclonal-monoclonal sandwich immunoassays, monoclonal -polyclonal sandwich
immunoassays, including radioisotope detection (radioimmunoassay (RIA)) and
enzyme
detection (enzyme immunoassay (EIA) or enzyme-linked immunosorbent assay
(ELISA)
(e.g., Quantikine ELISA assays, R&D Systems, Minneapolis, MN)). An example of
a point-
of-care device that can be used is i-STATS (Abbott, Laboratories, Abbott Park,
IL). Other
methods that can be used include a chemiluminescent microparticle immunoassay,
in
particular one employing the ARCHITECT automated analyzer (Abbott
Laboratories,
Abbott Park, IL), is an example. Other methods include, for example, mass
spectrometry,
and immunohistochemistry (e.g., with sections from tissue biopsies), using
anti-analyte (e.g.,
anti-UCH-L1) antibodies (monoclonal, polyclonal, chimeric, humanized, human,
etc.) or
antibody fragments thereof against analyte (e.g., UCH-L1). Other methods of
detection
include those described in, for example, U.S. Patent Nos. 6,143,576;
6,113,855; 6,019,944;
5,985,579; 5,947,124; 5,939,272; 5,922,615; 5,885,527; 5,851,776; 5,824,799;
5,679,526;
5,525,524; and 5,480,792.
Specific immunological binding of the antibody to the analyte (e.g., UCH-L1)
can be
detected via direct labels, such as fluorescent or luminescent tags, metals
and radionuclides
attached to the antibody or via indirect labels, such as alkaline phosphatase
or horseradish
peroxidase.
[0247] The use of immobilized antibodies or antibody fragments thereof may be
incorporated
into the immunoassay. The antibodies may be immobilized onto a variety of
supports, such
as magnetic or chromatographic matrix particles, the surface of an assay plate
(such as
microtiter wells), pieces of a solid substrate material, and the like. An
assay strip can be
prepared by coating the antibody or plurality of antibodies in an array on a
solid support.
This strip can then be dipped into the test sample and processed quickly
through washes and
detection steps to generate a measurable signal, such as a colored spot.
[0248] A homogeneous format may be used. For example, after the test sample
is obtained
from a subject, a mixture is prepared. The mixture contains the test sample
being assessed
for analyte (e.g., UCH-L1), a first specific binding partner, and a second
specific binding
89
Date Recue/Date Received 2023-06-29

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
partner. The order in which the test sample, the first specific binding
partner, and the second
specific binding partner are added to form the mixture is not critical. The
test sample is
simultaneously contacted with the first specific binding partner and the
second specific
binding partner. In some embodiments, the first specific binding partner and
any UCH-L1
contained in the test sample may form a first specific binding partner-analyte
(e.g., UCH-
L1)-antigen complex and the second specific binding partner may form a first
specific
binding partner-analyte of interest (e.g., UCH-L1)-second specific binding
partner complex.
In some embodiments, the second specific binding partner and any UCH-L1
contained in the
test sample may foim a second specific binding partner-analyte (e.g., UCH-L1)-
antigen
complex and the first specific binding partner may form a first specific
binding partner-
analyte of interest (e.g., UCH-L1)-second specific binding partner complex.
The first
specific binding partner may be an anti-analyte antibody (e.g., anti-UCH-L1
antibody that
binds to an epitope having an amino acid sequence comprising at least three
contiguous (3)
amino acids of SEQ ID NO: 1). The second specific binding partner may be an
anti-analyte
antibody (e.g., anti-UCH-L1 antibody that binds to an epitope having an amino
acid sequence
comprising at least three contiguous (3) amino acids of SEQ ID NO: 1).
Moreover, the
second specific binding partner is labeled with or contains a detectable label
as described
above.
[0249] A heterogeneous format may be used. For example, after the test
sample is obtained
from a subject, a first mixture is prepared. The mixture contains the test
sample being
assessed for analyte (e.g., UCH-L1) and a first specific binding partner,
wherein the first
specific binding partner and any UCH-L1 contained in the test sample form a
first specific
binding partner-analyte (e.g., UCH-L1)-antigen complex. The first specific
binding partner
may be an anti-analyte antibody (e.g., anti-UCH-L1 antibody that binds to an
epitope having
an amino acid sequence comprising at least three contiguous (3) amino acids of
SEQ ID NO:
1). The order in which the test sample and the first specific binding partner
are added to
form the mixture is not critical.
[0250] The first specific binding partner may be immobilized on a solid
phase. The solid
phase used in the immunoassay (for the first specific binding partner and,
optionally, the
second specific binding partner) can be any solid phase known in the art, such
as, but not

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
limited to, a magnetic particle, a bead, a test tube, a microtiter plate, a
cuvette, a membrane, a
scaffolding molecule, a film, a filter paper, a disc, and a chip. In those
embodiments where
the solid phase is a bead, the bead may be a magnetic bead or a magnetic
particle. Magnetic
beads/particles may be ferromagnetic, ferrimagnetic, paramagnetic,
superparamagnetic or
ferrofluidic. Exemplary ferromagnetic materials include Fe, Co, Ni, Gd, Dy,
Cr02, MnAs,
MnBi, Eu0, and NiO/Fe. Examples of ferrimagnetic materials include NiFe204,
CoFe204,
Fe304 (or Fe0Fe203). Beads can have a solid core portion that is magnetic and
is surrounded
by one or more non-magnetic layers. Alternately, the magnetic portion can be a
layer around
a non-magnetic core. The solid support on which the first specific binding
member is
immobilized may be stored in dry form or in a liquid. The magnetic beads may
be subjected
to a magnetic field prior to or after contacting with the sample with a
magnetic bead on
which the first specific binding member is immobilized.
[0251] After the mixture containing the first specific binding partner-
analyte (e.g., UCH-L1)
antigen complex is formed, any unbound analyte (e.g., UCH-L1) is removed from
the
complex using any technique known in the art. For example, the unbound analyte
(e.g.,
UCH-LI) can be removed by washing. Desirably, however, the first specific
binding partner
is present in excess of any analyte (e.g., UCH-L1) present in the test sample,
such that all
analyte (e.g., UCH-Li) that is present in the test sample is bound by the
first specific binding
partner.
[0252] After any unbound analyte (e.g., UCH-LI) is removed, a second
specific binding
partner is added to the mixture to form a first specific binding partner-
analyte of interest
(e.g., UCH-L1)-second specific binding partner complex. The second specific
binding
partner may be an anti-analyte antibody (e.g., anti-UCH-LI antibody that binds
to an epitope
having an amino acid sequence comprising at least three contiguous (3) amino
acids of SEQ
ID NO: 1). Moreover, the second specific binding partner is labeled with or
contains a
detectable label as described above.
[0253] The use of immobilized antibodies or antibody fragments thereof may
be incorporated
into the immunoassay. The antibodies may be immobilized onto a variety of
supports, such
as magnetic or chromatographic matrix particles (such as a magnetic bead),
latex particles or
modified surface latex particles, polymer or polymer film, plastic or plastic
film, planar
91

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
substrate, the surface of an assay plate (such as microtiter wells), pieces of
a solid substrate
material, and the like. An assay strip can be prepared by coating the antibody
or plurality of
antibodies in an array on a solid support. This strip can then be dipped into
the test sample
and processed quickly through washes and detection steps to generate a
measurable signal,
such as a colored spot.
(1) Sandwich immunoassay
[0254] A sandwich immunoassay measures the amount of antigen between two
layers of
antibodies (i.e., at least one capture antibody) and a detection antibody
(i.e., at least one
detection antibody). The capture antibody and the detection antibody bind to
different
epitopes on the antigen, e.g., analyte of interest such as UCH-Li. Desirably,
binding of the
capture antibody to an epitope does not interfere with binding of the
detection antibody to an
epitope. Either monoclonal or polyclonal antibodies may be used as the capture
and
detection antibodies in the sandwich immunoassay.
[0255] Generally, at least two antibodies are employed to separate and
quantify analyte (e.g.,
UCH-L1) in a test sample. More specifically, the at least two antibodies bind
to certain
epitopes of analyte (e.g., UCH-L1) forming an immune complex which is referred
to as a
"sandwich". One or more antibodies can be used to capture the analyte (e.g.,
UCH-L1) in the
test sample (these antibodies are frequently referred to as a "capture"
antibody or "capture"
antibodies) and one or more antibodies is used to bind a detectable (namely,
quantifiable)
label to the sandwich (these antibodies are frequently referred to as the
"detection" antibody
or "detection" antibodies). In a sandwich assay, the binding of an antibody to
its epitope
desirably is not diminished by the binding of any other antibody in the assay
to its respective
epitope. Antibodies are selected so that the one or more first antibodies
brought into contact
with a test sample suspected of containing analyte (e.g., UCH-L1) do not bind
to all or part of
an epitope recognized by the second or subsequent antibodies, thereby
interfering with the
ability of the one or more second detection antibodies to bind to the analyte
(e.g., UCH-L1).
[0256] The antibodies may be used as a first antibody in said immunoassay.
The antibody
immunospecifically binds to epitopes on analyte (e.g., UCH-L1). In addition to
the
antibodies of the present disclosure, said immunoassay may comprise a second
antibody that
immunospecifically binds to epitopes that are not recognized or bound by the
first antibody.
92

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0257] A test sample suspected of containing analyte (e.g., UCH-Li) can be
contacted with at
least one first capture antibody (or antibodies) and at least one second
detection antibodies
either simultaneously or sequentially. In the sandwich assay format, a test
sample suspected
of containing analyte (e.g., UCH-L1) is first brought into contact with the at
least one first
capture antibody that specifically binds to a particular epitope under
conditions which allow
the formation of a first antibody-analyte (e.g., UCH-L1) antigen complex. If
more than one
capture antibody is used, a first multiple capture antibody-UCH-L1 antigen
complex is
formed. In a sandwich assay, the antibodies, preferably, the at least one
capture antibody, are
used in molar excess amounts of the maximum amount of analyte (e.g., UCH-L1)
expected
in the test sample. For example, from about 5 g/m1 to about 1 mg/ml of
antibody per ml of
microparticle coating buffer may be used.
1. Anti-UCH-L1 Capture Antibody
[0258] Optionally, prior to contacting the test sample with the at least
one first capture
antibody, the at least one first capture antibody can be bound to a solid
support which
facilitates the separation the first antibody-analyte (e.g., UCH-L1) complex
from the test
sample. Any solid support known in the art can be used, including but not
limited to, solid
supports made out of polymeric materials in the forms of wells, tubes, or
beads (such as a
microparticle). The antibody (or antibodies) can be bound to the solid support
by adsorption,
by covalent bonding using a chemical coupling agent or by other means known in
the art,
provided that such binding does not interfere with the ability of the antibody
to bind analyte
(e.g., UCH-L1). Moreover, if necessary, the solid support can be derivatized
to allow
reactivity with various functional groups on the antibody. Such derivatization
requires the
use of certain coupling agents such as, but not limited to, maleic anhydride,
N-
hydroxysuccinimide and 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide.
[0259] After the test sample suspected of containing analyte (e.g., UCH-L1)
is incubated in
order to allow for the formation of a first capture antibody (or multiple
antibody)-analyte
(e.g., UCH-L1) complex. The incubation can be carried out at a pH of from
about 4.5 to
about 10.0, at a temperature of from about 2 C to about 45 C, and for a period
from at least
about one (1) minute to about eighteen (18) hours, from about 2-6 minutes,
from about 7 -12
minutes, from about 5-15 minutes, or from about 3-4 minutes.
93

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
it. Detection Antibody
[0260] After formation of the first/multiple capture antibody-analyte
(e.g., UCH-L1) complex,
the complex is then contacted with at least one second detection antibody
(under conditions
that allow for the formation of a first/multiple antibody-analyte (e.g., UCH-
L1) antigen-
second antibody complex). In some embodiments, the test sample is contacted
with the
detection antibody simultaneously with the capture antibody. If the first
antibody-analyte
(e.g., UCH-Li) complex is contacted with more than one detection antibody,
then a
first/multiple capture antibody-analyte UCH-L1)-multiple antibody detection
complex
is formed. As with first antibody, when the at least second (and subsequent)
antibody is
brought into contact with the first antibody-analyte (e.g., UCH-L1) complex, a
period of
incubation under conditions similar to those described above is required for
the formation of
the first/multiple antibody-analyte (e.g., UCH-L1)-second/multiple antibody
complex.
Preferably, at least one second antibody contains a detectable label. The
detectable label can
be bound to the at least one second antibody prior to, simultaneously with or
after the
formation of the first/multiple antibody-analyte (e.g., UCH-L1)-
second/multiple antibody
complex. Any detectable label known in the art can be used.
102611 Chemiluminescent assays can be performed in accordance with the
methods described
in Adamczyk et al., Anal. Chim. Acta 579(1): 61-67 (2006). While any suitable
assay format
can be used, a microplate chemiluminometer (Mithras LB-940, Berthold
Technologies
U.S.A., LLC, Oak Ridge, TN) enables the assay of multiple samples of small
volumes
rapidly. The chemiluminometer can be equipped with multiple reagent injectors
using 96-
well black polystyrene microplates (Costar #3792). Each sample can be added
into a
separate well, followed by the simultaneous/sequential addition of other
reagents as
determined by the type of assay employed. Desirably, the formation of
pseudobases in
neutral or basic solutions employing an acridinium aryl ester is avoided, such
as by
acidification. The chemiluminescent response is then recorded well-by-well. In
this regard,
the time for recording the chemiluminescent response will depend, in part, on
the delay
between the addition of the reagents and the particular acridinium employed.
[0262] The order in which the test sample and the specific binding
partner(s) are added to
form the mixture for chemiluminescent assay is not critical. If the first
specific binding
94

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
partner is detectably labeled with an acridinium compound, detectably labeled
first specific
binding partner-UCH-L1 antigen complexes form. Alternatively, if a second
specific binding
partner is used and the second specific binding partner is detectably labeled
with an
acridinium compound, detectably labeled first specific binding partner-analyte
(e.g., UCH-
L1)-second specific binding partner complexes form, Any unbound specific
binding partner,
whether labeled or unlabeled, can be removed from the mixture using any
technique known
in the art, such as washing.
[0263] Hydrogen peroxide can be generated in situ in the mixture or
provided or supplied to
the mixture before, simultaneously with, or after the addition of an above-
described
acridinium compound. Hydrogen peroxide can be generated in situ in a number of
ways such
as would be apparent to one skilled in the art.
[0264] Alternatively, a source of hydrogen peroxide can be simply added to
the mixture. For
example, the source of the hydrogen peroxide can be one or more buffers or
other solutions
that are known to contain hydrogen peroxide. In this regard, a solution of
hydrogen peroxide
can simply be added.
[0265] Upon the simultaneous or subsequent addition of at least one basic
solution to the
sample, a detectable signal, namely, a chemiluminescent signal, indicative of
the presence of
UCH-L1 is generated. The basic solution contains at least one base and has a
pH greater than
or equal to 10, preferably, greater than or equal to 12. Examples of basic
solutions include,
but are not limited to, sodium hydroxide, potassium hydroxide, calcium
hydroxide,
ammonium hydroxide, magnesium hydroxide, sodium carbonate, sodium bicarbonate,
calcium hydroxide, calcium carbonate, and calcium bicarbonate. The amount of
basic
solution added to the sample depends on the concentration of the basic
solution. Based on
the concentration of the basic solution used, one skilled in the art can
easily determine the
amount of basic solution to add to the sample. Other labels other than
chemiluminescent
labels can be employed. For instance, enzymatic labels (including but not
limited to alkaline
phosphatase) can be employed.
[0266] The chemiluminescent signal, or other signal, that is generated can
be detected using
routine techniques known to those skilled in the art. Based on the intensity
of the signal
generated, the amount of analyte of interest (e.g., UCH-L1) in the sample can
be quantified,

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
Specifically, the amount of analyte (e.g., UCH-L1) in the sample is
proportional to the
intensity of the signal generated. The amount of analyte (e.g., UCH-Li)
present can be
quantified by comparing the amount of light generated to a standard curve for
analyte (e.g.,
UCH-L1) or by comparison to a reference standard. The standard curve can be
generated
using serial dilutions or solutions of known concentrations of analyte (e.g.,
UCH-L1) by
mass spectroscopy, gravimetric methods, and other techniques known in the art.
(2) Forward Competitive Inhibition Assay
[0267] In a forward competitive format, an aliquot of labeled analyte of
interest (e.g., analyte
(e.g., UCH-L1) having a fluorescent label, a tag attached with a cleavable
linker, etc.) of a
known concentration is used to compete with analyte of interest (e.g., UCH-L1)
in a test
sample for binding to analyte of interest antibody (e.g., UCH-L1 antibody).
[0268] In a forward competition assay, an immobilized specific binding
partner (such as an
antibody) can either be sequentially or simultaneously contacted with the test
sample and a
labeled analyte of interest, analyte of interest fragment or analyte of
interest variant thereof.
The analyte of interest peptide, analyte of interest fragment or analyte of
interest variant can
be labeled with any detectable label, including a detectable label comprised
of tag attached
with a cleavable linker. In this assay, the antibody can be immobilized on to
a solid support.
Alternatively, the antibody can be coupled to an antibody, such as an
antispecies antibody,
that has been immobilized on a solid support, such as a microparticle or
planar substrate.
[0269] The labeled analyte of interest, the test sample and the antibody
are incubated under
conditions similar to those described above in connection with the sandwich
assay format.
Two different species of antibody-analyte of interest complexes may then be
generated.
Specifically, one of the antibody-analyte of interest complexes generated
contains a
detectable label (e.g., a fluorescent label, etc.) while the other antibody-
analyte of interest
complex does not contain a detectable label. The antibody-analyte of interest
complex can
be, but does not have to be, separated from the remainder of the test sample
prior to
quantification of the detectable label. Regardless of whether the antibody-
analyte of interest
complex is separated from the remainder of the test sample, the amount of
detectable label in
the antibody-analyte of interest complex is then quantified. The concentration
of analyte of
interest (such as membrane-associated analyte of interest, soluble analyte of
interest,
96

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
fragments of soluble analyte of interest, variants of analyte of interest
(membrane-associated
or soluble analyte of interest) or any combinations thereof) in the test
sample can then be
determined, e.g., as described above.
(3) Reverse Competitive Inhibition Assay
[0270] In a reverse competition assay, an immobilized analyte of interest
(e.g., UCH-L1) can
either be sequentially or simultaneously contacted with a test sample and at
least one labeled
antibody.
[0271] The analyte of interest can be bound to a solid support, such as the
solid supports
discussed above in connection with the sandwich assay format.
[0272] The immobilized analyte of interest, test sample and at least one
labeled antibody are
incubated under conditions similar to those described above in connection with
the sandwich
assay format. Two different species analyte of interest-antibody complexes are
then
generated. Specifically, one of the analyte of interest-antibody complexes
generated is
immobilized and contains a detectable label (e.g., a fluorescent label, etc.)
while the other
analyte of interest-antibody complex is not immobilized and contains a
detectable label. The
non-immobilized analyte of interest-antibody complex and the remainder of the
test sample
are removed from the presence of the immobilized analyte of interest-antibody
complex
through techniques known in the art, such as washing. Once the non-immobilized
analyte of
interest antibody complex is removed, the amount of detectable label in the
immobilized
analyte of interest-antibody complex is then quantified following cleavage of
the tag. The
concentration of analyte of interest in the test sample can then be determined
by comparing
the quantity of detectable label as described above.
(4) One-Step Immunoassay or "Capture on the Fly" Assay
[0273] In a capture on the fly immunoassay, a solid substrate is pre-coated
with an
immobilization agent. The capture agent, the analyte (e.g., UCH-L1) and the
detection agent
are added to the solid substrate together, followed by a wash step prior to
detection. The
capture agent can bind the analyte (e.g., UCH-L1) and comprises a ligand for
an
immobilization agent. The capture agent and the detection agents may be
antibodies or any
other moiety capable of capture or detection as described herein or known in
the art. The
ligand may comprise a peptide tag and an immobilization agent may comprise an
anti-
97

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
peptide tag antibody. Alternately, the ligand and the immobilization agent may
be any pair
of agents capable of binding together so as to be employed for a capture on
the fly assay
(e.g., specific binding pair, and others such as are known in the art). More
than one analyte
may be measured. In some embodiments, the solid substrate may be coated with
an antigen
and the analyte to be analyzed is an antibody.
[0274] In certain other embodiments, in a one-step immunoassay or "capture
on the fly", a
solid support (such as a microparticle) pre-coated with an immobilization
agent (such as
biotin, streptavidin, etc.) and at least a first specific binding member and a
second specific
binding member (which function as capture and detection reagents,
respectively) are used.
The first specific binding member comprises a ligand for the immobilization
agent (for
example, if the immobilization agent on the solid support is streptavidin, the
ligand on the
first specific binding member may be biotin) and also binds to the analyte of
interest (e.g.,
UCH-L1). The second specific binding member comprises a detectable label and
binds to an
analyte of interest (e.g., UCH-Li). The solid support and the first and second
specific
binding members may be added to a test sample (either sequentially or
simultaneously). The
ligand on the first specific binding member binds to the immobilization agent
on the solid
support to form a solid support/first specific binding member complex. Any
analyte of
interest present in the sample binds to the solid support/first specific
binding member
complex to form a solid support/first specific binding member/analyte complex.
The second
specific binding member binds to the solid support/first specific binding
member/analyte
complex and the detectable label is detected. An optional wash step may be
employed before
the detection. In certain embodiments, in a one-step assay more than one
analyte may be
measured. In certain other embodiments, more than two specific binding members
can be
employed. In certain other embodiments, multiple detectable labels can be
added. In certain
other embodiments, multiple analytes of interest can be detected, or their
amounts, levels or
concentrations, measured, determined or assessed.
102751 The use of a capture on the fly assay can be done in a variety of
fol mats as described
herein, and known in the art. For example, the format can be a sandwich assay
such as
described above, but alternately can be a competition assay, can employ a
single specific
binding member, or use other variations such as are known.
98

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
14. Kits
[0276] Provided herein is a kit, which may be used for obtaining a blood
test sample from a
subject and assaying or assessing the test sample for levels of UCH-L1 or UCH-
LI fragment.
The kits of the present disclosure can include a package of components for
drawing a blood
sample. Without limitation, kits can be used for obtaining blood samples from,
for example,
an adult patient, a juvenile patient, a neonatal patient, and a patient that
may have suffered a
traumatic brain injury. Components of the kit can include a means for
sterilizing the subject's
skin in the area of intended puncture. A typical and conventional sterilizing
means is a piece
of fabric commonly referred to as a gauze. The gauze preferably has a
sterilant occluded
therein and optionally may be attached to a grasping portion. A preferred
sterilant is alcohol,
although other sterilants such as antibacterial agents may be used. Exemplary
of suitable
antibacterials are the bisbiguanides, of which chlorhexidine is the best known
member. The
chlorhexidine may preferably be provided as a soluble salt in aqueous or
alcoholic solution.
[0277] The skin penetrating means for initiation of blood flow may be a
conventional lancet
or may be any of a variety of conventional devices which includes a needle.
The needle may
be single or double ended and may be of any gauge, preferably 21 or 23 gauge.
The needle
may include a safety sleeve, may be attached to a needle hub, and can be used
with a
conventional tube holder. The needle may also be part of a conventional
syringe assembly
including barrel and plunger. Also, as known in the art, the penetrating means
may be part of
a conventional blood collection set in which a penetrating needle having a
grasping means,
such as wings, is connected via a hub and tubing to a delivery needle for
puncture of a
septum of an evacuated tube.
[0278] The holding means of the kit may be any type of container for
receiving the sample,
such as, for example, a syringe barrel. Preferred holding means are
conventional tubes or
vials having a closed end and an open end. Such tubes may have an internal
volume of 100 ill
to 10 mL. Smaller tubes are generally used with a lancet for collection of
very small
quantities of blood by gravity flow. Representative of such tubes are
MicrotainerTM brand
tubes supplied by Becton, Dickinson and Company. The tube of the kit may also
be an
evacuated tube in which the open end is covered by a puncturable septum or
stopper, such as
VacutainerTM brand tubes supplied by Becton Dickinson and Co. Evacuated tubes
are
99

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
generally used with a conventional tube holder and blood collection set for
collection of
multiple larger blood samples, and may contain any of a variety of
conventional blood
analysis additives, such as anticoagulants, blood clotting agents and serum
separating gels.
Exemplary anticoagulants can include lithium heparin and ethylenediaminetetra
acetic acid
(EDTA), and can include a siliceous particle. The holding means may also be a
test strip in
which the sample is allowed to flow directly onto a glass or plastic strip
containing reagents
for analysis, such as by immunoassay.
[0279] When a subject's skin is punctured in order to take a blood sample,
the puncture
wound is usually covered to stanch bleeding and protect the wound during
healing.
Accordingly, the kit of the invention may include additional gauze to cover
the wound and a
means, such as adhesive, to affix the covering gauze over the wound. In
accordance with
current medical practice, the kit can include a disposable glove to avoid any
contact between
the phlebotomist and the sample. To promote blood flow, the kit may also
include a
tourniquet.
[0280] All components of the kit may be supplied in packaging, such as a
compartmentalized
plastic enclosure, preferably with a hermetically sealable cover so that the
contents of the kit
can be sterilized and sealed for storage.
[0281] The kit can include at least one component for assaying the test
sample for UCH-Li
instructions for assaying the test sample for UCH-Li. For example, the kit can
comprise
instructions for assaying the test sample for UCH-L1 by immunoassay, e.g.,
chemiluminescent microparticle immunoassay. Instructions included in kits can
be affixed to
packaging material or can be included as a package insert. While the
instructions are
typically written or printed materials they are not limited to such. Any
medium capable of
storing such instructions and communicating them to an end user is
contemplated by this
disclosure. Such media include, but are not limited to, electronic storage
media (e.g.,
magnetic discs, tapes, cartridges, chips), optical media (e.g., CD ROM), and
the like. As used
herein, the term "instructions" can include the address of an internet site
that provides the
instructions. The at least one component may include at least one composition
comprising
one or more isolated antibodies or antibody fragments thereof that
specifically bind to UCH-
Ll. The antibody may be an UCH-L1 capture antibody and/or a UCH-L1 detection
antibody.
100

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0282] Alternatively or additionally, the kit can comprise a calibrator or
control, e.g., purified,
and optionally lyophilized, UCH-L1, and/or at least one container (e.g., tube,
microtiter
plates or strips, which can be already coated with an anti-UCH-L1 monoclonal
antibody) for
conducting the assay, and/or a buffer, such as an assay buffer or a wash
buffer, either one of
which can be provided as a concentrated solution, a substrate solution for the
detectable label
(e.g., an enzymatic label), or a stop solution. Preferably, the kit comprises
all components,
i.e., reagents, standards, buffers, diluents, etc., which are necessary to
perform the assay. The
instructions also can include instructions for generating a standard curve.
[0283] The kit may further comprise reference standards for quantifying UCH-
Ll. The
reference standards may be employed to establish standard curves for
interpolation and/or
extrapolation of UCH-Li concentrations. The reference standards may include a
high UCH-
Li concentration level, for example, about 100000 pg/mL, about 125000 pg/mL,
about
150000 pg/mL, about 175000 pg/mL, about 200000 pg/mL, about 225000 pg/mL,
about
250000 pg/mL, about 275000 pg/mL, or about 300000 pg/mL; a medium UCH-L1
concentration level, for example, about 25000 pg/mL, about 40000 pg/mL, about
45000
pg/mL, about 50000 pg/mL, about 55000 pg/mL, about 60000 pg/mL, about 75000
pg/mL or
about 100000 pg/mL; and/or a low UCH-L1 concentration level, for example,
about 1
pg/mL, about 5 pg/mL, about 10 pg/mL, about 12.5 pg/mL, about 15 pg/mL, about
20
pg/mL, about 25 pg/mL, about 30 pg/mL, about 35 pg/mL, about 40 pg/mL, about
45 pg/mL,
about 50 pg/mL, about 55 pg/mL, about 60 pg/mL, about 65 pg/mL, about 70
pg/mL, about
75 pg/mL, about 80 pg/mL, about 85 pg/mL, about 90 pg/mL, about 95 pg/mL, or
about 100
pg/mL.
[0284] Any antibodies, which are provided in the kit, such as recombinant
antibodies specific
for UCH-L1, can incorporate a detectable label, such as a fluorophore,
radioactive moiety,
enzyme, biotin/avidin label, chromophore, chemiluminescent label, or the like,
or the kit can
include reagents for labeling the antibodies or reagents for detecting the
antibodies (e.g.,
detection antibodies) and/or for labeling the analytes (e.g., UCH-L1) or
reagents for detecting
the analyte (e.g., UCH-L1). The antibodies, calibrators, and/or controls can
be provided in
separate containers or pre-dispensed into an appropriate assay foiinat, for
example, into
microtiter plates.
101

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0285] Optionally, the kit includes quality control components (for
example, sensitivity
panels, calibrators, and positive controls). Preparation of quality control
reagents is well-
known in the art and is described on insert sheets for a variety of
immunodiagnostic products.
Sensitivity panel members optionally are used to establish assay performance
characteristics,
and further optionally are useful indicators of the integrity of the
immunoassay kit reagents,
and the standardization of assays,
[0286] The kit can also optionally include other reagents required to
conduct a diagnostic
assay or facilitate quality control evaluations, such as buffers, salts,
enzymes, enzyme co-
factors, substrates, detection reagents, and the like. Other components, such
as buffers and
solutions for the isolation and/or treatment of a test sample (e.g.,
pretreatment reagents), also
can be included in the kit. The kit can additionally include one or more other
controls. One
or more of the components of the kit can be lyophilized, in which case the kit
can further
comprise reagents suitable for the reconstitution of the lyophilized
components.
[0287] The various components of the kit optionally are provided in
suitable containers as
necessary, e.g., a microtiter plate. The kit can further include containers
for holding or
storing a sample (e.g., a container or cartridge for a urine, whole blood,
plasma, or serum
sample). Where appropriate, the kit optionally also can contain reaction
vessels, mixing
vessels, and other components that facilitate the preparation of reagents or
the test sample.
The kit can also include one or more instrument for assisting with obtaining a
test sample,
such as a syringe, pipette, forceps, measured spoon, or the like.
[0288] If the detectable label is at least one acridinium compound, the kit
can comprise at
least one acridinium-9-carboxamide, at least one acridinium-9-carboxylate aryl
ester, or any
combination thereof. If the detectable label is at least one acridinium
compound, the kit also
can comprise a source of hydrogen peroxide, such as a buffer, solution, and/or
at least one
basic solution. If desired, the kit can contain a solid phase, such as a
magnetic particle, bead,
test tube, microtiter plate, cuvette, membrane, scaffolding molecule, film,
filter paper, disc,
or chip.
[0289] If desired, the kit can further comprise one or more components,
alone or in further
combination with instructions, for assaying the test sample for another
analyte, which can be
a biomarker, such as a biomarker of traumatic brain injury or disorder.
102

[0290] The kit (or components thereof), as well as the method for assessing
or determining
the concentration of UCH-L1 in a test sample by an immunoassay as described
herein, can be
adapted for use in a variety of automated and semi-automated systems
(including those
wherein the solid phase comprises a microparticle), as described, e.g., U.S.
Patent No.
5,063,081, U.S. Patent Application Publication Nos. 2003/0170881,
2004/0018577,
2005/0054078, and 2006/0160164 and as commercially marketed e.g., by Abbott
Laboratories (Abbott Park, IL) as Abbott Point of Care (i-STATil or i-STAT
Alinity, Abbott
Laboratories) as well as those described in U.S. Patent Nos. 5,089,424 and
5,006,309, and as
commercially marketed, e.g., by Abbott Laboratories (Abbott Park, IL) as
ARCHITECT or
the series of Abbott Alinity devices.
[0291] Some of the differences between an automated or semi-automated system
as compared
to a non-automated system (e.g., ELISA) include the substrate to which the
first specific
binding partner (e.g., analyte antibody or capture antibody) is attached
(which can affect
sandwich formation and analyte reactivity), and the length and timing of the
capture,
detection, and/or any optional wash steps. Whereas a non-automated format such
as an
ELISA may require a relatively longer incubation time with sample and capture
reagent (e.g.,
about 2 hours), an automated or semi-automated format (e.g., ARCHITECT and
any
successor platform, Abbott Laboratories) may have a relatively shorter
incubation time (e.g.,
approximately 18 minutes for ARCHITECT ). Similarly, whereas a non-automated
format
such as an ELISA may incubate a detection antibody such as the conjugate
reagent for a
relatively longer incubation time (e.g., about 2 hours), an automated or semi-
automated
format (e.g., ARCHITECTS and any successor platform) may have a relatively
shorter
incubation time (e.g., approximately 4 minutes for the ARCHITECT = and any
successor
platform).
[0292] Other platforms available from Abbott Laboratories include, but are
not limited to,
AxSYM -4, IMx (see, e.g., U.S. Patent No. 5,294,404),
PRISM , E1A (bead), and QuantumTM IL as well as other
platforms. Additionally, the assays, kits, and kit components can be employed
in other
formats, for example, on electrochemical or other hand-held or point-of-care
assay systems.
As mentioned previously, the present disclosure is, for example, applicable to
the
103
Date Recue/Date Received 2023-06-29

commercial Abbott Point of Care (i-STAT1, Abbott Laboratories) electrochemical
immunoassay system that performs sandwich immunoassays. Immunosensors and
their
methods of manufacture and operation in single-use test devices are described,
for example
in, U.S. Patent No. 5,063,081, U.S. Patent App. Publication Nos. 2003/0170881,
2004/0018577, 2005/0054078, and 2006/0160164.
102931 In particular, with regard to the adaptation of an assay to the i-
STAT system, the
following configuration is preferred. A microfabricated silicon chip is
manufactured with a
pair of gold amperometric working electrodes and a silver-silver chloride
reference electrode.
On one of the working electrodes, polystyrene beads (0.2 pm diameter) with
immobilized
capture antibody are adhered to a polymer coating of patterned polyvinyl
alcohol over the
electrode. This chip is assembled into an i-STAT cartridge with a fluidics
format suitable
for immunoassay. On a portion of the silicon chip, there is a specific binding
partner for
UCH-L1, such as one or more UCH-L1 antibodies (one or more
monoclonal/polyclonal
antibody or a fragment thereof, a variant thereof, or a fragment of a variant
thereof that can
bind UCH-L1) or one or more anti-UCH-L1 DVD-Igs (or a fragment thereof, a
variant
thereof, or a fragment of a variant thereof that can bind UCH-L1), either of
which can be
detectably labeled. Within the fluid pouch of the cartridge is an aqueous
reagent that
includes p-aminophenol phosphate.
102941 In operation, a sample from a subject suspected of suffering from
TBI is added to the
holding chamber of the test cartridge, and the cartridge is inserted into the
i-STATe reader.
A pump element within the cartridge pushes the sample into a conduit
containing the chip.
The sample is brought into contact with the sensors allowing the enzyme
conjugate to
dissolve into the sample. The sample is oscillated across the sensors to
promote formation of
the sandwich of approximately 2-12 minutes. In the penultimate step of the
assay, the sample
is pushed into a waste chamber and wash fluid, containing a substrate for the
alkaline
phosphatase enzyme, is used to wash excess enzyme conjugate and sample off the
sensor
chip. In the final step of the assay, the alkaline phosphatase label reacts
with p-aminophenol
phosphate to cleave the phosphate group and permit the liberated p-aminophenol
to be
electrochemically oxidized at the working electrode. Based on the measured
current, the
104
Date Recue/Date Received 2023-06-29

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
reader is able to calculate the amount of UCH-L1 in the sample by means of an
embedded
algorithm and factory-determined calibration curve.
[0295] The methods and kits as described herein necessarily encompass other
reagents and
methods for carrying out the immunoassay. For instance, encompassed are
various buffers
such as are known in the art and/or which can be readily prepared or optimized
to be
employed, e.g., for washing, as a conjugate diluent, and/or as a calibrator
diluent. An
exemplary conjugate diluent is ARCHITECT conjugate diluent employed in
certain kits
(Abbott Laboratories, Abbott Park, IL) and containing 2-(N-
morpholino)ethanesulfonic acid
(MES), a salt, a protein blocker, an antimicrobial agent, and a detergent. An
exemplary
calibrator diluent is ARCHITECT human calibrator diluent employed in certain
kits
(Abbott Laboratories, Abbott Park, IL), which comprises a buffer containing
IVIES, other salt,
a protein blocker, and an antimicrobial agent. Additionally, as described in
U.S. Patent
Application No. 61/142,048 filed December 31, 2008, improved signal generation
may be
obtained, e.g., in an i-STAT cartridge format, using a nucleic acid sequence
linked to the
signal antibody as a signal amplifier.
[0296] While certain embodiments herein are advantageous when employed to
assess disease,
such as traumatic brain injury, the assays and kits also optionally can be
employed to assess
UCH-L1 in other diseases, disorders, and conditions as appropriate.
[0297] The method of assay also can be used to identify a compound that
ameliorates
diseases, such as traumatic brain injury. For example, a cell that expresses
UCH-L1 can be
contacted with a candidate compound. The level of expression of UCH-L1 in the
cell
contacted with the compound can be compared to that in a control cell using
the method of
assay described herein.
15. Examples
10298] It will be readily apparent to those skilled in the art that other
suitable modifications
and adaptations of the methods of the present disclosure described herein are
readily
applicable and appreciable, and may be made using suitable equivalents without
departing
from the scope of the present disclosure or the aspects and embodiments
disclosed
herein. Having now described the present disclosure in detail, the same will
be more clearly
105

understood by reference to the following examples, which are merely intended
only to
illustrate some aspects and embodiments of the disclosure, and should not be
viewed as
limiting to the scope of the disclosure.
[0299] The present disclosure has multiple aspects, illustrated by the
following non-limiting
examples.
Example 1
i-STAT UCH-L1 Assay
[0300] An i-STAT UCH-LI assay in development was used in the examples
described herein.
However, it is understood that any UCH-L1 assay using the same or different
antibodies can
be improved using the present disclosure described herein. Monoclonal antibody
pairs, such
as Antibody A as a capture monoclonal antibody and Antibody B and C as a
detection
monoclonal antibody, were used. Antibody A is an exemplary anti-UCH-L1
antibody that
was internally developed at Abbott Laboratories (Abbott Park, IL). Antibody B
and C,
developed by Banyan Biomarkers (Alachua, Florida), recognize different
epitopes of UCH-
Li and enhance the detection of antigen in the sample. Other antibodies that
were internally
developed at Abbott Laboratories (Abbott Park, IL) or which have been
described in the
literature also show or are expected to show similar enhancement of signal
when used
together as capture antibodies or detection antibodies, in various
combinations. The UCH-L1
assay design was evaluated against key performance attributes. The cartridge
configuration
was Antibody Configuration: Antibody A (Capture Antibody)/Antibody B+C
(Detection
Antibody); Reagent conditions: 0.8% solids, 125 pig /mL Fab Alkaline
Phosphatase cluster
conjugate; and Sample Inlet Print: UCH-L1 standard. The assay time was 10-15
mm (with 7-
12 min sample capture time).
Example 2
Stability of UCH-L1 in Whole Blood
[0301] The stability of UCH-L1 in blood samples was evaluated due to the
observation that
subjects that have not experienced an injury to the head, or have not been
diagnosed as
106
Date Recue/Date Received 2023-06-29

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
having a disease or disorder relating to UCH-L1 ("normal" subjects), have
measurable levels
of UCH-L1 in whole blood samples. The i-STAT platform was used to measure UCH-
L1
in whole blood without prior separation of the cells from the plasma portion,
and the results
are shown in FIGS. 1A-1B.
[0302] The initial experiment was designed to understand the stability of
the endogenous
UCH-L1 antigen in whole blood from normal subjects. Blood was drawn from 5
normal
donors into EDTA collection tubes following an approved Institutional Review
Board study
protocol; each participant gave written informed consent. The blood was
continuously mixed
at room temperature for 24 hours and tested at 1 hour, 4 hour, 8 hour and 24
hour time points
after collection/mixing. A small amount of the blood was transferred into an
Eppendorfrm
LoBind tube at each time point and centrifuged to test the corresponding
plasma at each time
point.
[0303] As demonstrated in FIGS. 1A-1B (UCH-L1 levels and UCH-L1 levels as a
percent of
baseline, respectively), the concentration of UCH-L1 for each of the whole
blood samples
increased at each time point and over 24 hours. Additionally, there was a
significant rise in
UCH-L1 concentration with each of the whole blood samples that were
continuously mixed
for the full 24 hours (e.g., meaning during the entire time during the testing
period). The
difference from the point when the experiment began for each donor (e.g.,
baseline (which
was the first testing time point once the whole blood was obtained) is shown
in FIG. 1B.
Example 3
Stability of UCH-L1 in Plasma
[0304] The stability of UCH-L1 in plasma samples was evaluated in normal
subjects (FIGS.
2A-2B), UCH-L1 concentration of the corresponding plasma separated at each of
these time
points discussed above in Example 1 (FIGS. 1A-1B) showed similar results.
Blood was
drawn from 5 normal donors into EDTA collection tubes following an approved
Institutional
Review Board study protocol; each participant gave written informed consent.
The blood was
continuously mixed at room temperature for 24 hours and tested at 1 hour, 4
hour, 8 hour and
24 hour time points after collection/mixing. A small amount of the blood was
transferred into
an Eppendorf TM LoBind tube at each time point and centrifuged to test the
corresponding
107

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
plasma at each time point. As shown in FIG. 2A, there was a 2 to 5 fold
increase in the
plasma values over 24 hours when the plasma was not separated from the whole
blood until
the sample was removed from the mixing device and processed for UCH-L1
assessment. The
continuous mixing used in these experiments indicates that hemolysis may be an
important
factor influencing UCH-L1 measurements.
[0305] As demonstrated in FIGS. 2A-2B (UCH-L1 levels and UCH-L1 levels as a
percent of
baseline, respectively), the concentration of UCH-L1 for each of the plasma
samples
increased at each time point and over 24 hours when stored as whole blood
during the time
period. Additionally, there was a significant rise in UCH-L1 concentration
with each of the
whole blood samples that were continuously mixed for the full 24 hours. The
difference from
the point when the experiment began for each donor (baseline) is shown in FIG.
2B.
Example 4
Effects of Anti-coagulants on UCH-L1 Stability
[0306] Experiments were conducted to investigate the effects of anti-
coagulants on the
stability of UCH-Li in whole blood samples from normal subjects. Blood was
collected in Li
heparin, EDTA and serum tubes from 4 donors. The EDTA and lithium (Li) heparin
whole
blood samples were quickly aliquoted into 2 mL tubes, and these samples were
held
stationary at room temperature and 2-8 C for 2 hour, 6 hour, 24 hours, and 48
hours before
processing and testing for UCH-L1 levels. At each time point, whole blood
samples (FIGS.
3A-3B) and the corresponding plasma samples (Table 2) were assessed for UCH-Li
concentrations. The whole blood samples were thoroughly mixed by inversion
(approximately ten times (which takes less 60 seconds)) prior to testing.
[0307] Stability of UCH-L1 from whole blood samples processed in Li heparin
tubes at room
temperature compared to 2-8 C is shown in FIG. 3A. UCH-L1 concentrations were
stable
(<10% change) in whole blood samples at room temperature for at least the
initial 24 hours
after the samples were obtained, when stored stationary. However, UCH-L1
concentrations
from whole blood samples from the same donors were only stable at 2-8 C until
about the
first 6 hours; there was at least about a 10% difference (increase in UCH-L1
levels) in three
of the four samples at 24 hours and 48 hours (FIG. 3A). See Table 2 below.
108

CA 03068041 2019-12-19
WO 2019/010131
PCT/US2018/040612
[0308] Stability of UCH-L1 from whole blood samples processed in EDTA tubes at
room
temperature compared to 2-8 C is shown in FIG. 3B. UCH-L1 concentrations were
stable
(<15% change) in whole blood samples at room temperature for at least the
initial 6 hours
after the samples were obtained, when stored stationary. However, UCH-L1
concentrations
from whole blood samples from the same donors tested at 24 hours and 48 hours
did show
that storage at 2-8 C improved UCH-L1 stability compared to storage at room
temperature.
There was less than about a 20% difference in the concentrations of samples
stored at 2-8 C,
whereas the whole blood stored at room temperature showed an increase in UCH-
Li
concentrations up to about 200% higher than the initial concentrations (FIG.
3B). See Table 2
below.
Table 2 Concentrations of UCH-L1 (pg/mL) for whole blood sample stability
testing.
Time
Tube
After Sample Donor 1 Donor 2
Donor 3 Donor 4
Type
Draw
Li Hep 154.7 55.0 143.8
80.6
Baseline Whole Blood
EDTA 154.7 66.9 149.6
89.2
RT Li Hep 156.8 51.0 144.6
80.5
2 h Whole EDTA 163.3 65.8 163.6
97.1
ours
Blood 2 8 C Li Hep 152.5 56.0 142.2
77.7
- EDTA 161.3 68.4 153.0
100.4
RT Li Hep 161.5 56.8 144.1
81.6
6 h Whole EDTA 164.5 71.9 163.6
98.2
ours
Blood 2 8 C Li Hep 162.0 53.5 151.8
84.4
- EDTA 166.2 66.4 162.1
100.5
RT Li Hep 156.4 53.1 155.8
86.9
24 h Whole EDTA 208.5 128.5 291.3
209.3
ours
Blood 2 8 C Li Hep 158.1 63.4 158.9
95.7
- EDTA 182.5 66.9 173.5
95.1
RT Li Hep 161.1 100.2 241.4
106.1
48 h Whole EDTA 302.3 177.3 458.1
273.1
ours
Blood 2-8 C Li Hep 161.9 83.4 219.4
99.6
EDTA 181.8 60.8 180.2
90.9
[0309]
Stability of UCH-L1 from corresponding plasma samples processed in EDTA and Li
heparin tubes at room temperature compared to 2-8 C is shown in Table 3
below. At each
time point for each of the whole blood samples tested, the corresponding
plasma samples
109

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
were isolated and immediately tested to evaluate the variability in the plasma
samples. The
UCH-L1 concentration values and the differences observed in the plasma samples
were very
similar to the results in whole blood (FIGS. 3A-3B), thus indicating that the
factors
contributing to the increased concentrations of UCH-L1 at 24 hours and 48
hours likely
involve the pre-analytical phases of sample processing (i.e., after obtaining
the blood samples
and/or during storage) and did not vary with the reagents.
Table 3 Concentrations of UCH-L1 (pg/mL) for plasma sample stability
testing.
Time
Tube
After Sample Donor 1 Donor 2 Donor 3
Donor 4
Type
Draw
Baseline Plasma Li Hep 113.1 28.2 80.2 56.8
EDTA 117.1 42.2 79.8 58.5
Li Hep 114.6 , 33.0 88.0
58.1
RT
EDTA 121.8 39.4 79.8 62.5
2 hours Plasma
Li Hep 124.3 43.0 94.5 59.6
2-8 C
EDTA 121.4 40.0 80.1 58.4
Li Hep 106.7 33.5 81.7 52.3
RT
EDTA 112.5 38.0 77.0 57.6
6 hours Plasma
Li Hep 123.9 36.4 106.0 67.8
2-8 C
EDTA 120.6 41.3 91.3 64.3
Li Hep 113.0 , 32.8 83.5
50.3
RT
EDTA 154.2 73.6 178.9 155.5
24 hours Plasma
Li Hep 117.4 44.6 101.6 73.7
2-8 C
EDTA 143.1 39.5 90.2 58.3
Li Hep 116.0 62.0 139.7 67.3
RT EDTA 253.2 118.3 , 341.2 , 230.5
48 hours Plasma
Li Hep , 122.3 69.6 146.9 81.5
2-8 C
EDTA 139.8 37.5 101.4 57.6
[0310] The results in Tables 2 and 3 show that Li heparin and EDTA tubes
show similar
effect as there is some donor-to-donor variability in both sample types (whole
blood and
plasma). However, the results in Tables 2 and 3 demonstrate an improvement in
the stability
of UCH-L1 levels in the sample when the amount of mixing is reduced by holding
the
sample stationary (namely, not continuously mixing as shown in Example 2)
during storage
regardless of the temperature (namely, room temperature or at 2-8 C). Further
improvement
in the stability of UCH-L1 levels in the sample is shown when such samples
(e.g., those that
110

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
are not continuously mixed) are stored at a temperature of 2-8 C (such samples
are stable up
to 24 to 48 hours after being obtained from a subject).
Example 5
Stability of UCH-L1 in Plasma and Serum
[0311] Experiments were conducted to assess the stability of UCH-L1 in plasma
and serum
samples that were isolated shortly after whole blood samples were obtained
from normal
donors, in contrast to being isolated after the whole blood samples were
subjected to mixing
and/or stored at room temperature or 2-8 C. Fresh serum and plasma was
separated prior to
initiation of the experiment (time point 0). The samples were then aliquoted
and stored at
room temperature or 2-8 C. At each time point (0 hours, 2 hours, 8 hours, 24
hours, and 48
hours after draw), an aliquot of plasma was mixed and tested for UCH-L1
levels.
[0312] As shown in FIGS. 4A (room temperature) and 4B (2-8 C), UCH-L1 was
stable for
just the initial few hours after obtaining the blood samples and separating
the plasma using
EDTA tubes, and there was no significant difference in UCH-L1 stability in
plasma at room
temperature compared to 2-8 C. In FIGS. 5A (room temperature) and 5B (2-8
C), the use of
Li heparin tubes did not produce significantly different results as compared
to EDTA tubes.
Additionally, as shown in FIGS. 6A (room temperature) and 6B (2-8 C), the use
of serum
tubes to isolate serum and test for UCH-L1 levels did not produce
significantly different
results as compared to plasma samples.
[0313] These results contrast with the results above for whole blood
samples, in which UCH-
Li concentrations increased at 24 and 48 hours, thus indicating that whole
blood samples
reacted differently than plasma and serum samples to the same processing
conditions.
Specifically, these results show that sample handling is important with
respect to whole
blood samples (e.g., mixing should be avoided with these samples and these
samples should
be stored at 2-8 C).
111

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
Example 6
Effects of Clot Formation on UCH-L1 Stability in Serum
[0314] Serum is the liquid portion of the whole blood that is isolated
after a clot and clotting
factors are folmed (i.e., plasma without the clotting factors). Generally, it
takes about 30-60
min for a clot to form in a whole blood sample at room temperature before the
samples can
be centrifuged to separate the serum. It is possible for cells to lyse during
the clotting
process, which may result in the release of cellular components in the serum.
To evaluate the
effects a clot may have on the components of serum when serum is exposed to a
clot, two
tubes of blood from 4 normal donors were collected in standard serum tubes.
The serum was
isolated from one tube and transferred to a secondary tube for each of the
donors. The
secondary tube was held stationary at room temperature for 8 hours, and serum
from the
second tube with the clot was separated after 8 hours and tested.
[0315] As shown in FIG. 7, UCH-L1 concentrations were higher when the serum
was not
separated from the clot. UCH-L1 concentrations were at least 30% higher for
each of the
samples, thus indicating the possibility of release of UCH-L1 from the clot
into the serum.
[0316] In another experiment, fresh serum and plasma were isolated from
whole blood within
1 hour of collection and aliquoted into separate Eppendorfrm LoBind tubes, and
each of the
aliquots were stored at RT or 2-8 C. The plasma and serum aliquots were
tested at 2 hours, 8
hours, 24 hours, and 48 hours to evaluate the stability of the neat plasma and
serum that was
isolated from the whole blood samples. Table 4 below shows the concentrations
of the
plasma sample for each of the 4 donors over time. UCH-L1 levels in the samples
were
relatively stable at 2-8 C for up to 8 hours, and at RT for up to 2 hours.
However, unlike the
whole blood stability results, the concentration of UCH-L1 decreased with time
at both
temperatures for all sample types, suggesting that the increase in UCH-Li
levels in the whole
blood samples was due to presence of the cellular components of the blood. In
summary,
these results show that sample handling is extremely important with respect to
whole blood
samples (e.g., mixing should be avoided with these samples and these samples
should be
stored at 2-8 C). In contrast, once plasma and serum are separated from
cells/clots, these
samples demonstrate good stability up to 8 hours.
112

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
Table 4 Concentrations of UCH-L1 (pg/mL) for sample stability testing.
Storage at RT
Concentration
Donor sample type
Time 0 2 hour 8 hour 24 hour 48 hour
1 serum 160.0 145.5 125.5 101.5 80.5
2 serum 46.8 43.9 38.2 25.7 33.4
3 serum 130.2 133.1 115.2 96.2 83.8
4 serum 80.6 72.3 65.6 49.6 40.3
1 , K2 EDTA , 153.4 135.9 126.1 118.4 117.0
,
2 K2 EDTA , 45.1 43.9 47.6 31.0 45.0
3 K2 EDTA 95.1 89,3 87.5 79.7 76.7
_
4 K2 EDTA 68.4 65,1 63.6 52.3 51.0
1 Li-Hep 155.1 139.5 134.6 91.3 95.7
2 Li-Hep 40.2 41.7 30.7 21.6 24.3
3 Li-Hep 106.6 104.2 102.0 76.0 75.3
4 Li-Hep 70.5 66.5 64.8 42.6 40.4
Storage at 2 to 8 C
Concentration
Donor sample type
, Time 0 2 hour 8 hour
24 hour , 48 hour ,
1 serum 160.0 155.8 139.5 , 104.2
111.5
2 serum 46.8 50.7 49.9 33.1 34.5
3 serum 130.2 135.3 132.1 99.1 95.5
4 serum 80,6 77.9 81.2 55.3 51.2
1 K2 EDTA 153.4 150.6 142.2 120.3 116.3
2 K2 EDTA 45.1 44.8 46.3 38.0 33.8
3 K2 EDTA 95.1 101.0 90.9 83.5 79.1
4 K2 EDTA 68.4 69.2 64.6 57.8 47.4
1 Li-Hep 155.1 146.7 149.4 94.6 99.6
2 Li-Hep 40.2 38.3 41.5 29.4 31.4
3 Li-Hep 106.6 102.5 103.3 80.1 81.4
4 Li-Hep 70.5 63.8 63.2 48.7 52.4
113

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
Example 7
Contribution of Hemolysis for UCH-L1 Stability
[0317] Evaluation of interference studies indicated the possibility of a
component present in
hemolysate as the cause of the increase in UCH-L1 readings, when hemolysate
was spiked
into a UCH-L1 low panel (data not shown). It was also shown that this UCH-L1
"interference" signal could be specifically reduced by addition of the anti-
UCH-Li capture
antibody used in the immunoassay; however, non-specific mouse IgG showed no
difference
when it was added as a control.
[0318] To test the possibility of hemolysate interference, four hemolysates
were prepared
using the packed red blood cells (RBC's) from the bottom of the tube from four
normal
donor subjects. Removing cells from the bottom was an attempt to minimize the
amount of
buffy coat in these hemolysates. The packed RBCs were washed with saline
repeatedly and
lysed by freezing to generate the hemolysate solution. A small portion of this
hemolysate
solution was added to heat treated EDTA plasma (containing approximately 10
pg/mL UCH-
L1), and tested to evaluate the change in signal.
[0319] Each hemolysate was spiked into an EDTA plasma pool targeting three
concentrations
of hemoglobin (0.75, 0.50, and 0.25 g/dL) and the samples were tested using
UCH-L1 assay.
The hemoglobin level for each of these samples was measured on a hematology
analyzer.
During the period of spiking and measurement, the sample was mixed for 20 to
30 minutes as
known in the art. FIG. 8 shows the UCH-L1 concentration differences as a
function of
hemoglobin concentration. Additionally, FIG. 9 shows that whole blood samples
from 3
normal donors spiked with recombinant UCH-L1 did not exhibit the same increase
in UCH-
Li over time. These data indicate that the increase in UCH-L1 levels in whole
blood
described above is more apparent in subjects with relatively low baseline UCH-
Li
concentrations.
[0320] It is understood that the foregoing detailed description and
accompanying examples
are merely illustrative and are not to be taken as limitations upon the scope
of the invention,
which is defined solely by the appended claims and their equivalents.
[0321] Various changes and modifications to the disclosed embodiments will
be apparent to
those skilled in the art. Such changes and modifications, including without
limitation those
114

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
relating to the chemical structures, substituents, derivatives, intermediates,
syntheses,
compositions, formulations, or methods of use of the invention, may be made
without
departing from the spirit and scope thereof.
[0322] For reasons of completeness, various aspects of the invention are
set out in the
following numbered clauses:
[0323] Clause 1. In an improvement of a method of measuring an amount of
ubiquitin
carboxy-terminal hydrolase Li (UCH-L1) in a whole blood sample obtained from a
subject,
wherein said improvement comprises processing the sample within no more than
about eight
hours after the sample is obtained from the subject to avoid a rise in UCH-L1
level that
results from storage of the sample.
[0324] Clause 2. The improvement of clause 1, wherein the sample is
processed within a
period of time after the sample is obtained from the subject selected from the
group
consisting of: (a) from about zero hours to about 6 hours; (b) from about zero
hours to about
4 hours; (c) from about zero hours to about 2 hours; and (d) from about zero
hours to about 1
hour.
[0325] Clause 3. The improvement of clause 1, wherein the sample is
processed within a
period of time after the sample is obtained from the subject selected from the
group
consisting of: (a) from about 1 hour to about 8 hours; (b) from about 1 hour
to about 6 hours;
(c) from about 1 hour to about 4 hours; and (d) from about 1 hour to about 2
hours.
[0326] Clause 4. The improvement of any one of clauses 1-3, wherein
processing the sample
comprises separating plasma from blood cells in the sample; and, subsequently
performing a
test using the plasma that measures the amount of UCH-Li.
[0327] Clause 5. The improvement of any one of clauses 1-4, wherein the
sample is obtained
from the subject using a container comprising an anticoagulant selected from
the group
consisting of heparin and EDTA.
[0328] Clause 6. The improvement of any one of clauses 1-4, wherein
processing the sample
comprises separating serum from any clots that arise in the sample; and,
subsequently
performing a test using the serum that measures the amount of UCH-Ll.
[0329] Clause 7. The improvement of clause 6, wherein the sample is
collected using a serum
collection tube.
115

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0330] Clause 8. The improvement of any one of clauses 1-3, wherein
processing the sample
comprises performing a test that measures the amount of UCH-LI in the sample.
[0331] Clause 9. The improvement of any one of clauses 4-8, wherein the
test comprises any
method by which UCH-L1 amount can be assessed.
[0332] Clause 10. The improvement of clause 9, wherein the test is selected
from the group
consisting of an immunoassay, chemical analysis, SDS PAGE and Western blot
analysis,
electrophoresis analysis, a protein assay, a competitive binding assay, a
functional protein
assay, chromatography, and spectrophotometry.
[0333] Clause 11. The improvement of any one of clauses 4-10, wherein the
test is employed
in a clinical chemistry format.
[0334] Clause 12. The improvement of clause 10 or 11, wherein the test is
an immunoassay
comprising: contacting the sample, either simultaneously or sequentially, in
any order with:
(1) at least one capture antibody, which binds to an epitope on UCH-L1 or UCH-
L1 fragment
to form a capture antibody-UCH-L1 antigen complex, and (2) at least one
detection antibody
which includes a detectable label and binds to an epitope on UCH-L1 that is
not bound by the
capture antibody, to form a UCH-L1 antigen-detection antibody complex, such
that a capture
antibody-UCH-Li antigen-detection antibody complex is formed, and measuring
the amount
or concentration of UCH-L1 in the sample based on the signal generated by the
detectable
label in the capture antibody-UCH-L1 antigen-detection antibody complex.
[0335] Clause 13. The improvement of any one of clauses 4-12, wherein the
sample is
maintained at room temperature for some period of time during the time point
that the sample
is obtained from the subject to a time point when the test is performed.
[0336] Clause 14. The improvement of any one of clauses 4-12, wherein the
sample is
maintained at a temperature from about 2 C to about 8 C for some period of
time during the
time point that the sample is obtained from the subject to the time point when
the test is
performed.
[0337] Clause 15. The improvement of any one of clauses 8-12, wherein the
sample is not
mixed between the period of time after the sample is obtained from the subject
to the time
point when the test is performed.
116

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0338] Clause 16. The improvement of any one of clauses 1-15, wherein the
UCH-L1
amount in the sample from the subject is assessed as a measure of traumatic
brain injury,
wherein the subject may have sustained an injury to the head.
[0339] Clause 17. A method of avoiding or preventing an increase or rise in
ubiquitin
carboxy-terminal hydrolase Ll (UCH-L1) levels between the period of time a
whole blood
sample is obtained from a subject and prior to performing an assay on the
sample, the method
comprising the step of: processing the sample within no more than about eight
hours after
the sample is obtained from the subject to avoid an increase or rise in UCH-L1
level that
results from storage of the sample prior to performing the assay.
[0340] Clause 18. The method of clause 17, wherein the method avoids or
prevents an
erroneously high level of UCH-L1 in the sample between the period of time the
sample is
obtained from the subject and prior to performing the assay
[0341] Clause 19. The method of clause 17, wherein the sample is processed
within a period
of time after the sample is obtained from the subject selected from the group
consisting of:
(a) from about zero hours to about 6 hours; (b) from about zero hours to about
4 hours; (c)
from about zero hours to about 2 hours; and (d) from about zero hours to about
1 hour.
[0342] Clause 20. The method of clause 17, wherein the sample is processed
within a period
of time after the sample is obtained from the subject selected from the group
consisting of:
(a) from about 1 hour to about 8 hours; (b) from about 1 hour to about 6
hours; (c) from
about 1 hour to about 4 hours; and (d) from about 1 hour to about 2 hours.
[0343] Clause 21. The method of any one of clauses 17-20, further
comprising subsequently
performing an assay to measure the amount of UCH-L1 in the sample.
[0344] Clause 22. The method of any one of clauses 17-20, wherein
processing the sample
comprises separating plasma from blood cells in the sample; and, subsequently
performing
an assay using the plasma that measures the amount of UCH-L1 in the sample.
[0345] Clause 23. The method of any one of clauses 17-22, wherein the
sample is obtained
from the subject using a container comprising an anticoagulant selected from
the group
consisting of heparin and EDTA.
117

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0346] Clause 24. The method of any one of clauses 17-22, wherein
processing the sample
comprises separating serum from any clots that arise in the sample; and,
subsequently
performing an assay using the serum that measures the amount of UCH-L1 in the
sample
[0347] Clause 25. The method of clause 24, wherein the sample is collected
using a serum
collection tube.
[0348] Clause 26. The method of any one of clauses 17-25, wherein the assay
comprises any
method by which UCH-L1 amount can be assessed.
[0349] Clause 27. The method of clause 26, wherein the assay is selected
from the group
consisting of an immunoassay, chemical analysis, SDS PAGE and Western blot
analysis,
electrophoresis analysis, a protein assay, a competitive binding assay, a
functional protein
assay, chromatography, and spectrophotometry.
[0350] Clause 28. The method of any one of clauses 21-26, wherein the assay
is employed in
a clinical chemistry format.
[0351] Clause 29. The method of clause 27, wherein the assay is an
immunoassay
comprising:
a) contacting the sample, either simultaneously or sequentially, in any order
with:
(1) at least one capture antibody, which binds to an epitope on UCH-L1 or UCH-
Li fragment to form a capture antibody-UCH-L1 antigen complex, and
(2) at least one detection antibody which includes a detectable label and
binds to
an epitope on UCH-L1 that is not bound by the capture antibody, to form a UCH-
Li antigen-detection antibody complex,
such that a capture antibody-UCH-L1 antigen-detection antibody complex is
formed, and
b) measuring the amount or concentration of UCH-L1 in the sample based on the
signal generated by the detectable label in the capture antibody-UCH-L1
antigen-
detection antibody complex.
[0352] Clause 30. The method of any one of clauses 21-29, wherein the
sample is maintained
at room temperature for some period of time between the time point that the
sample is
obtained from the subject and the time point when the assay is performed.
118

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0353] Clause 31. The method of any one of clauses 21-29, wherein the
sample is maintained
at a temperature from about 2 C to about 8 C for some period of time between
the time
point that the sample is obtained from the subject and the time point when the
assay is
performed.
[0354] Clause 32. The method of any one of clauses 25-29, wherein the
sample is not mixed
between the period of time between the time point that the sample is obtained
from the
subject and the time point when the test is performed.
[0355] Clause 33. The method of any one of clauses 17-32, wherein the UCH-
L1 amount in
the sample from the subject is assessed as a measure of traumatic brain
injury, wherein the
subject may have sustained an injury to the head.
[0356] Clause 34. In an improvement of a method of measuring an amount of
ubiquitin
carboxy-terminal hydrolase Ll (UCH-L1) in a whole blood sample obtained from a
subject,
wherein said improvement comprises reducing errors in UCH-L1 measurements by
controlling preanalyti cal processing conditions.
[0357] Clause 35. The improvement of clause 34, wherein controlling
preanalytical
processing conditions comprise processing the sample within on more than about
eight hours
after the sample is obtained from the subject.
[0358] Clause 36. The improvement of clause 35, wherein the sample is
processed within a
period of time after the sample is obtained from the subject selected from the
group
consisting of: (a) from about zero hours to about 6 hours; (b) from about zero
hours to about
4 hours; (c) from about zero hours to about 2 hours; and (d) from about zero
hours to about 1
hour.
[0359] Clause 37. The improvement of clause 35, wherein the sample is
processed within a
period of time after the sample is obtained from the subject selected from the
group
consisting of: (a) from about 1 hour to about 8 hours; (b) from about 1 hour
to about 6 hours;
(c) from about 1 hour to about 4 hours; and (d) from about 1 hour to about 2
hours.
[0360] Clause 38. The improvement of any one of clauses 35-37, wherein
processing the
sample comprises separating plasma from blood cells in the sample; and,
subsequently
performing a test using the plasma that measures the amount of UCH-Ll.
119

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0361] Clause 39. The improvement of any one of clauses 34-38, wherein the
sample is
obtained from the subject using a container comprising an anticoagulant
selected from the
group consisting of heparin and EDTA.
[0362] Clause 40. The improvement of any one of clauses 35-39, wherein
processing the
sample comprises separating serum from any clots that arise in the sample;
and, subsequently
performing a test using the serum that measures the amount of UCH-Ll.
[0363] Clause 41. The improvement of clause 40, wherein the sample is
collected using a
serum collection tube.
[0364] Clause 42. The improvement of any one of clauses 35-40, wherein
processing the
sample comprises performing a test that measures the amount of UCH-L1 in the
sample.
[0365] Clause 43. The improvement of any one of clauses 38-41, wherein the
test comprises
any method by which UCH-L1 amount can be assessed.
[0366] Clause 44. The improvement of clause 43, wherein the test is
selected from the group
consisting of an immunoassay, chemical analysis, SDS PAGE and Western blot
analysis,
electrophoresis analysis, a protein assay, a competitive binding assay, a
functional protein
assay, chromatography, and spectrophotometry.
[0367] Clause 45. The improvement of any one of clauses 38-44, wherein the
test is
employed in a clinical chemistry format.
[0368] Clause 46. The improvement of clause 44 or 45, wherein the test is
an immunoassay
comprising: contacting the sample, either simultaneously or sequentially, in
any order with:
(1) at least one capture antibody, which binds to an epitope on UCH-L1 or UCH-
L1 fragment
to form a capture antibody-UCH-L1 antigen complex, and (2) at least one
detection antibody
which includes a detectable label and binds to an epitope on UCH-L1 that is
not bound by the
capture antibody, to form a UCH-L1 antigen-detection antibody complex, such
that a capture
antibody-UCH-L1 antigen-detection antibody complex is foimed, and measuring
the amount
or concentration of UCH-L1 in the sample based on the signal generated by the
detectable
label in the capture antibody-UCH-Li antigen-detection antibody complex.
[0369] Clause 47. The improvement of any one of clauses 38-46, wherein the
sample is
maintained at room temperature for some period of time during the time point
that the sample
is obtained from the subject to a time point when the test is performed.
120

CA 03068041 2019-12-19
WO 2019/010131 PCT/US2018/040612
[0370] Clause 48. The improvement of any one of clauses 38-46, wherein the
sample is
maintained at a temperature from about 2 C to about 8 C for some period of
time during the
time point that the sample is obtained from the subject to the time point when
the test is
performed.
[0371] Clause 49. The improvement of any one of clauses 42-46, wherein the
sample is not
mixed between the period of time after the sample is obtained from the subject
to the time
point when the test is perfoimed.
[0372] Clause 50. The improvement of any one of clauses 34-49, wherein the UCH-
L1
amount in the sample from the subject is assessed as a measure of traumatic
brain injury,
wherein the subject may have sustained an injury to the head.
121

Dessin représentatif

Désolé, le dessin représentatif concernant le document de brevet no 3068041 est introuvable.

États administratifs

2024-08-01 : Dans le cadre de la transition vers les Brevets de nouvelle génération (BNG), la base de données sur les brevets canadiens (BDBC) contient désormais un Historique d'événement plus détaillé, qui reproduit le Journal des événements de notre nouvelle solution interne.

Veuillez noter que les événements débutant par « Inactive : » se réfèrent à des événements qui ne sont plus utilisés dans notre nouvelle solution interne.

Pour une meilleure compréhension de l'état de la demande ou brevet qui figure sur cette page, la rubrique Mise en garde , et les descriptions de Brevet , Historique d'événement , Taxes périodiques et Historique des paiements devraient être consultées.

Historique d'événement

Description Date
Lettre envoyée 2024-06-25
Inactive : Octroit téléchargé 2024-06-25
Inactive : Octroit téléchargé 2024-06-25
Accordé par délivrance 2024-06-25
Inactive : Page couverture publiée 2024-06-24
Préoctroi 2024-05-13
Inactive : Taxe finale reçue 2024-05-13
month 2024-01-22
Lettre envoyée 2024-01-22
Un avis d'acceptation est envoyé 2024-01-22
Inactive : Approuvée aux fins d'acceptation (AFA) 2023-12-22
Inactive : Q2 réussi 2023-12-22
Modification reçue - réponse à une demande de l'examinateur 2023-06-29
Modification reçue - modification volontaire 2023-06-29
Lettre envoyée 2023-05-25
Exigences de prorogation de délai pour l'accomplissement d'un acte - jugée conforme 2023-05-25
Demande de prorogation de délai pour l'accomplissement d'un acte reçue 2023-04-28
Rapport d'examen 2022-12-29
Inactive : Rapport - Aucun CQ 2022-12-19
Lettre envoyée 2022-02-18
Exigences pour une requête d'examen - jugée conforme 2022-01-20
Toutes les exigences pour l'examen - jugée conforme 2022-01-20
Requête d'examen reçue 2022-01-20
Représentant commun nommé 2020-11-07
Inactive : COVID 19 - Délai prolongé 2020-06-10
Inactive : Page couverture publiée 2020-02-06
Lettre envoyée 2020-01-22
Demande reçue - PCT 2020-01-17
Exigences applicables à la revendication de priorité - jugée conforme 2020-01-17
Demande de priorité reçue 2020-01-17
Inactive : CIB attribuée 2020-01-17
Inactive : CIB en 1re position 2020-01-17
LSB vérifié - pas défectueux 2019-12-20
Exigences pour l'entrée dans la phase nationale - jugée conforme 2019-12-19
Inactive : Listage des séquences à télécharger 2019-12-19
Inactive : Listage des séquences - Reçu 2019-12-19
Demande publiée (accessible au public) 2019-01-10

Historique d'abandonnement

Il n'y a pas d'historique d'abandonnement

Taxes périodiques

Le dernier paiement a été reçu le 2024-06-14

Avis : Si le paiement en totalité n'a pas été reçu au plus tard à la date indiquée, une taxe supplémentaire peut être imposée, soit une des taxes suivantes :

  • taxe de rétablissement ;
  • taxe pour paiement en souffrance ; ou
  • taxe additionnelle pour le renversement d'une péremption réputée.

Les taxes sur les brevets sont ajustées au 1er janvier de chaque année. Les montants ci-dessus sont les montants actuels s'ils sont reçus au plus tard le 31 décembre de l'année en cours.
Veuillez vous référer à la page web des taxes sur les brevets de l'OPIC pour voir tous les montants actuels des taxes.

Historique des taxes

Type de taxes Anniversaire Échéance Date payée
Taxe nationale de base - générale 2019-12-19 2019-12-19
TM (demande, 2e anniv.) - générale 02 2020-07-02 2020-06-18
TM (demande, 3e anniv.) - générale 03 2021-07-02 2021-06-16
Requête d'examen - générale 2023-07-04 2022-01-20
TM (demande, 4e anniv.) - générale 04 2022-07-04 2022-06-15
Prorogation de délai 2023-04-28 2023-04-28
TM (demande, 5e anniv.) - générale 05 2023-07-04 2023-06-14
Pages excédentaires (taxe finale) 2024-05-13 2024-05-13
Taxe finale - générale 2024-05-13
TM (demande, 6e anniv.) - générale 06 2024-07-02 2024-06-14
Titulaires au dossier

Les titulaires actuels et antérieures au dossier sont affichés en ordre alphabétique.

Titulaires actuels au dossier
ABBOTT LABORATORIES
Titulaires antérieures au dossier
DAVID PACENTI
GANGAMANI S. BELIGERE
JESSICA GRIESHABER
JOHN RAMP
MELISSA B. BRENNAN
SAUL A. DATWYLER
Les propriétaires antérieurs qui ne figurent pas dans la liste des « Propriétaires au dossier » apparaîtront dans d'autres documents au dossier.
Documents

Pour visionner les fichiers sélectionnés, entrer le code reCAPTCHA :



Pour visualiser une image, cliquer sur un lien dans la colonne description du document (Temporairement non-disponible). Pour télécharger l'image (les images), cliquer l'une ou plusieurs cases à cocher dans la première colonne et ensuite cliquer sur le bouton "Télécharger sélection en format PDF (archive Zip)" ou le bouton "Télécharger sélection (en un fichier PDF fusionné)".

Liste des documents de brevet publiés et non publiés sur la BDBC .

Si vous avez des difficultés à accéder au contenu, veuillez communiquer avec le Centre de services à la clientèle au 1-866-997-1936, ou envoyer un courriel au Centre de service à la clientèle de l'OPIC.


Description du
Document 
Date
(yyyy-mm-dd) 
Nombre de pages   Taille de l'image (Ko) 
Page couverture 2024-05-23 1 35
Revendications 2023-06-28 4 215
Description 2023-06-28 121 9 500
Description 2019-12-18 121 6 645
Dessins 2019-12-18 15 695
Abrégé 2019-12-18 1 62
Revendications 2019-12-18 5 207
Page couverture 2020-02-05 1 33
Paiement de taxe périodique 2024-06-13 24 989
Certificat électronique d'octroi 2024-06-24 1 2 528
Taxe finale 2024-05-12 4 94
Courtoisie - Lettre confirmant l'entrée en phase nationale en vertu du PCT 2020-01-21 1 594
Courtoisie - Réception de la requête d'examen 2022-02-17 1 424
Avis du commissaire - Demande jugée acceptable 2024-01-21 1 580
Modification / réponse à un rapport 2023-06-28 44 3 269
Rapport de recherche internationale 2019-12-18 3 83
Demande d'entrée en phase nationale 2019-12-18 3 78
Poursuite - Modification 2019-12-19 2 53
Requête d'examen 2022-01-19 3 82
Demande de l'examinateur 2022-12-28 5 316
Prorogation de délai pour examen 2023-04-27 4 103
Courtoisie - Demande de prolongation du délai - Conforme 2023-05-24 2 242

Listes de séquence biologique

Sélectionner une soumission LSB et cliquer sur le bouton "Télécharger la LSB" pour télécharger le fichier.

Si vous avez des difficultés à accéder au contenu, veuillez communiquer avec le Centre de services à la clientèle au 1-866-997-1936, ou envoyer un courriel au Centre de service à la clientèle de l'OPIC.

Soyez avisé que les fichiers avec les extensions .pep et .seq qui ont été créés par l'OPIC comme fichier de travail peuvent être incomplets et ne doivent pas être considérés comme étant des communications officielles.

Fichiers LSB

Pour visionner les fichiers sélectionnés, entrer le code reCAPTCHA :